Language selection

Search

Patent 2493799 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2493799
(54) English Title: LINCOMYCIN DERIVATIVES POSSESSING ANTIBACTERIAL ACTIVITY
(54) French Title: DERIVES DE LINCOMYCINE PRESENTANT UNE ACTIVITE ANTIBACTERIENNE
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07H 15/16 (2006.01)
  • A61K 31/7052 (2006.01)
  • A61P 31/04 (2006.01)
  • C07H 15/12 (2006.01)
(72) Inventors :
  • LEWIS, JASON (United States of America)
  • PATEL, DINESH V. (United States of America)
  • ANANDAN, SAMPATH K. (United States of America)
  • GORDEEV, MIKHAIL F. (United States of America)
(73) Owners :
  • VICURON PHARMACEUTICALS, INC. (United States of America)
(71) Applicants :
  • VICURON PHARMACEUTICALS, INC. (United States of America)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2003-08-15
(87) Open to Public Inspection: 2004-02-26
Examination requested: 2008-08-06
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2003/025820
(87) International Publication Number: WO2004/016632
(85) National Entry: 2005-01-26

(30) Application Priority Data:
Application No. Country/Territory Date
60/403,770 United States of America 2002-08-15

Abstracts

English Abstract




Novel lincomycin derivatives are disclosed. These lincomycin derivatives
exhibit antibacterial activity. As the compounds of the subject invention
exhibit potent activities against bacteria, including gram positive organisms,
they are useful antimicrobial agents. Methods of synthesis and of use of the
compounds are also disclosed.


French Abstract

L'invention concerne de nouveaux dérivés de lincomycine. Ces dérivés de lincomycine présentent une activité antibactérienne. Les composés de l'invention sont utiles en tant qu'agents antimicrobiens car ils présentent des activités puissantes contre les bactéries, y compris les organismes Gram positifs. L'invention concerne également des méthodes de synthèse et d'utilisation de ces composés.

Claims

Note: Claims are shown in the official language in which they were submitted.



CLAIMS

1. A compound of formula (I):
Image
wherein
R1 is alkyl;
R2 and R3 are independently H, alkyl, hydroxy, fluoro, or cyanoalkyl or
one, of R2 and R3 is =NOR7 and the other is absent, or one of R2 and R3 is
=CH2 and
the other is absent, with, the provisos that both R2 and R3 are not H; when
one of R2
and R3 is fluoro, the other is not hydrogen or hydroxy; and when one of R2 and
R3
is hydroxy, the other is not fluoro, hydrogen, or hydroxy;
R6 is selected from the groin consisting of H, alkyl,
hydroxyalkyl, -C(O)O-alkylene-cycloalkyl, -C(O)O-alkylene-substituted
cycloalkyl, -C(O)O-alkyl, -C(O)O-substituted
alkyl, -C(O)O-aryl, -C(O)O-substituted
aryl, -C(O)O-heteroaryl, -C(O)O-substituted
heteroaryl, -[C(O)O]p-alkylene-heterocycle, -[C(O)O]p-alkylene-substituted
heterocycle, wherein p is 0 or 1 with the proviso that -C(O)O-substituted
alkyl does
not include the following:

143





Image

R7 is H or alkyl;
R9, which can, be singly or multiply substituted in the ring on the same
or different carbons, is independently selected from the group consisting of
hydrogen, alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted
cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH2)n-OH, -(CH2)n-NR4R5, alkylene-R a where R a is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and R5 are H or alkyl;
and
m is 1 or 2; and
prodrugs, tautomers or pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula I has a minimum inhibition
coucentration of 32 µg/ml, or less against at least one of the organisms
selected from the
group consisting of Streptococcus pneumoniae, Staphylococcus aureus,
Staphylococcus
epidermidis, Enterococcus faecalis, Enterococcus faecium, Haemophilus
influenzae,
Moraxella catarrhalis, Escherichia coli, Bacteroides, fragilis, Bacteroides
thetaiotaomicron,
and Clostridium difficile.

2. A compound of formula (II)

Image


144


wherein:
R1 is alkyl;
R2 and R3 are independently H, alkyl, or cyanoalkyl, with the proviso
that both R2 and R3 are not H;
R6 is H, alkyl, or hydroxyalkyl;
R9, which can be singly or multiply substituted in the ring on the same
or different carbons, is independently selected from the group consisting of
hydrogen, alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted
cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH2)n-OH, -(CH2)n-NR4R5, -alkylene-R a where R a is selected from,
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and R5 are H or alkyl;
and
m is 1 or 2; and
prodrugs and pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula II has a minimum inhibition
concentration of 32 µg/mL or less against at least one of the organisms
selected from the
group consisting of Streptococcus pneumoniae, Staphylococcus aureus,
Staphylococcus
epidermidis, Enterococcus faecalis, Enterococcus faecium, Haemophilus
influenzae,
Moraxella catarrhalis, Escherichia coli, Bacteroides fragilis, Bacteroides
thetaiotaomicron,
and Clostridium difficile.

3. A compound of formula (III):

Image



145



wherein:
R1 is aryl;
R2 and R3 are fluoro;
R6 is H, alkyl, or hydroxyalkyl;
R9, which can be singly or multiply substituted in the ring on the same
or different carbons, is independently selected from the group consisting of
hydrogen, alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted
cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH2)n-OH, -(CH2)n-NR4R5, alkylene-R a where R a is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n as an integer of from 1 to 8 inclusive and R4 and R5 are H or alkyl;
and
m is 1 or 2; and
prodrugs and pharmaceutically acceptable salts thereof,
with the proviso that the compound of formula III has a minimum inhibition
concentration of 32 µg/mL or less against at least one of the organisms
selected from the
group consisting of Streptococcus pneumoniae, Staphylococcus aureus,
Staphylococcus
epidermidis, Enterococcus faecalis, Enterococcus faecium, Haemophilus
influenzae,
Moraxella catarrhalis, Escherichia coli, Bacteroides fragilis, Bacteroides
thetaiotaomicron,
and Clostridium difficile.

4. A compound of formula (IV):

Image

wherein:
R1 is alkyl;


146


R2 and R3 are independently H, or alkyl, hydroxy, fluoro, or cyanoalkyl
ox one of R2 and R3 is =NOR7 and the other is absent, or one of R2 and R3 is
=CH2
and the other is absent, with the provisos that both R2 and R3 are not H; when
one
of R2 and R3 is fluoro, the other is not hydrogen or hydroxy; and when one of
R2
and R3 is hydroxy, the other is not fluoro, hydrogen, or hydroxy;
R6 is selected from the group consisting
of -C(O)O-alkylene-cycloalkyl, -C(O)O-alkylene-substituted
cycloalkyl, -C(O)O-alkyl, -C(O)O-substituted
alkyl, -C(O)O-aryl, -C(O)O-substituted
aryl, -C(O)O-heteroaryl, -C(O)O-substituted
heteroaryl, -[C(O)O]p alkylene-heterocycle, -[C(O)O]p-alkylene-substituted
heterocycle, wherein p is 0 or 1 with the proviso that -C(O)O-substituted
alkyl does
not include the following:

Image


R7 is H or alkyl;
R9, which can be singly or multiply substituted in the ring on the same
or different carbons, is independently selected from the group consisting of
hydrogen, alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted
cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH2)n-OH, -(CH2)n-NR4R5, -alkylene-R a where R a is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and R5 are H or alkyl;
and
m is 1 or 2; and
prodrugs, tautomers or pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula I has a minimum,
inhibition concentration of 32 µg/mL or less against at least one of the


147


organisms selected from the group consisting of Streptococcus pneumoniae,
Staphylococcus aureus, Staphylococcus epidermidis, Enterococcus faecalis,
Enterococcus faecium, Haemophilus influenzae, Moraxella catarrhalis,
Escherichia coli, Bacteroides fragilis, Bacteroides thetaiotaomicron, and
Clostridium difficile.

5. A, compound of claim 1, wherein R9 is H, alkyl, ar substituted
alkyl.

6. A compound of claim 1, wherein R1 is methyl.

7. A compound selected from the group consisting of
4-methoxy-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(4,4-difluoro-butyl)-piperidine-2-carboxylic acid [2 methyl-1-
(3,4,5 trihydroxy-6-methylsulfanyl-tetrahydro pyran-2-yl)-propyl]-amide;
4-ethyl-piperidine 2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl tetrahydro-pyran-2-yl)-propyl]-amide;
1-methyl-4-propyl-pyrrolidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro pyran 2-yl) propyl-amide;
1-methyl-4-propyl-pyrrolidine-2-carboxylic acid [3-cyano-2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran 2-yl)-propyl]-amide;
4-ethyl-piperidine-2-carboxylic acid [2 hydroxy-2-methyl-1,(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
1-methyl-4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxyimino-1-
(3,4,5 trihydroxy-6-methylsulfanyl-tetrahydro pyran-2-yl)-propyl]-amide;
1-methyl-4-propyl-pyrrolidine-2-carboxylic acid [2-methoxyimino-1-
(3,4,5 trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
5-butyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro pyran-2-yl)propyl]-amide;
4-pentyl-pyrrolidine-2-carboxylic acid (2methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3-methyl-butyl)-pyrrolidine-2-carboxylic acid [z-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl-propyl]-amide;



148



4-pentyl-pyrrolidine 2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
1-methyl-4-propyl-pyrrolidine-2-carboxylic acid [2,2-difluoro-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-pentyl-pyrrolidine-2-carboxylic acid [2,2-difluoro-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran 2-yl)-propyl]-amide;
4-[3-(4-fluoro phenyl)-propyl]-pyrrolidine-2-carboxylic acid [2-methyl-
1-(3,4,5 trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl) propyl]-amide;
4-(3,3-difluoro-propyl)-pyrrolidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy 6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-[3-(4-chloro-phenyl)-propyl]-pyrrolidine-2-carboxylic acid [2-methyl-
1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4(2,2-difluoro pentyl)-pyrrolidine 2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-propyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
1-(2-hydroxy-ethyl)-4-pentyl-pyrrolidine-2-carboxylic-acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl tetrahydro-pyran-2-yl)-propyl]-amide;
1-1(2-hydroxy propyl)-4-pentyl-pyrrolidine 2-carboxylic acid [2-methyl-
1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
1-(3-hydroxy-propyl)-4-pentyl-pyrrolidine-2-carboxylic acid [2-methyl-
1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran 2-yl)-propyl]-amide;
1-(2 hydroxy-ethyl)-(3 methyl-butyl)-pyrrolidine-2-carboxylic acid
[2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-
amide;
4-(3,3-dihydro-propyl)-1-(2-hydroxy-ethyl)-pyrrolidine-2-carboxylic
acid [2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl-
propyl]-amide;
1-(2-hydroxy-ethyl)-4-pentyl-pyrrolidine-2-carboxylic acid [2,2-
difluoro-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-
amide;
4-pentyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl tetrahydro-pyran-2-yl)-propyl]-amide;



149


4-(1-ethyl-propyl)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl)-amide;
4-isopropyl-piperidine-2-carboxylic acid (2-methyl-1-(3,4,5-trihydroxy
6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-butyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5 trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-cyclohexyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl tetrahydro pyran-2-yl)-propyl]-amide;
4-ethyl-1-(2-hydroxy-ethyl)-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl tetrahydro-pyran-2-yl)-propyl]-amide;
1-(2-hydroxy-ethyl)-4-pentyl-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
1-(2-hydroxy-ethyl)-4-propyl-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
2-[2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-
yl)-propylcarbamoyl]-4-propyl-piperidine-1-carboxylic acid 9H-fluoren-9-yl
methyl ester;
2-[2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-
yl)-propylcarbamoyl]-4-propyl-piperidine-1-carboxylic acid ethyl ester;
2-[2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-
yl)-propylcarbamoyl]-4-propyl-piperidine-1-carboxylic acid phenyl ester;
4-(4,4-difluoro-pentyl)-pyrrolidine-2-carboxylic acid [2-methyl-1-
(3,4,5-tridroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl-propyl]-amide;
4-(3,3-difluoro-butyl)-pyrrolidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3,3-difluoro-pentyl)-pyrrolidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihdroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3,3-difluoro-pentyl)-1-(2 hydroxy-ethyl)-pyrrolidine-2-carboxylic
acid [2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-
propyl]-amide;
4-(2,2-difluoro-ethyl)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3,3-difluoro-propyl)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;

150



4-(5,5-difluoro-pentyl))-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(5-fluoro-pentyl)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-5-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(4-fluoro-butyl)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihdroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3-ethyl-3-hydroxy-pentyl)-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-butoxy-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-pentyloxy-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-
6-methylsulfanyl-tetraydro-pyran-2-yl)-propyl]-amide;
4-(4-fluoro-butoxy)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-butyl-pyrrolidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-allyl]-amide;
1;4-diethyl-piperidine-2-carboxylic acid (2-methyl-1-(3,4,5-trihydroxy-
6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3-fluoro-propoxy)-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihdroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-(3,3,3-trifluoro-propoxy)-piperidine-2-carboxylic acid [2-methyl-1-
(3,4,5-trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-isobutyl-piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-propyl-piperidine-2-carboxylic acid [2,2-difluoro-1-(3,4,5-trihydroxy-
6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-fluoro-4-propyl-pyrrolidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-butyl-4-fluoro-pyrrolidine-2-carboxylic acid [2-methyl-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-fluoro-4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxy-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
and prodrugs, tautomers and pharmaceutically acceptable salts thereof.

151





8. A pharmaceutical composition comprising a pharmaceutically
acceptable carrier and a therapeutically effective amount of a compound of
any one of claims 1 - 7

9. A method for the treatment of a microbial infection in a
mammal comprising administering to the mammal a therapeutically effective
amount of a compound of any one of claims 1 - 7.

10. The method according to claim 9, wherein the compound is
administered to the mammal orally, parenterally, transdermally, topically,
rectally, or intranasally in a pharmaceutical composition.

11. The method according to claim 9, wherein the compound is
administered in an amount of from about 0.1 to about 100 mg/kg of body
weight/day.

12. A compound according to claim 7, wherein said compound is 4-propyl-
piperidine-2-carboxylic acid [2-methyl-1-(3,4,5-trihydroxy-6-methylsulfanyl-
tetrahydro-
pyran-2-yl)-propyl]-amide.

13. A compound having the structure
Image

14. A pharmaceutical composition comprising a pharmaceutically acceptable
carrier
and a therapeutically effective amount of the compound of claim 13.

15. A method for the treatment of a microbial infection in a mammal,
comprising
administering to the mammal a therapeutically effective amount of the compound
of claim
13.



152




16. A method for the treatment of a microbial infection in a mammal,
comprising
administering to the mammal a pharmaceutical composition according to claim
14.

17. A compound according to claim 1, wherein one of R2 and R3 is hydrogen, and
another of R2 and R3 is methyl.



153

Description

Note: Descriptions are shown in the official language in which they were submitted.




CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
LINCOMYCIN DERIVATIVES POSSESSING ANTIBACTERIAL ACTIVITY
[0001] This application claims the benefit under 35 U.S.C. ~119(e) of U.S.
Provisional
Application No. 60/403,770 filed on August 15, 2002, the disclosure of which
is hereby
incorporated by reference.
BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION
[0002] This invention relates to lincomycin derivatives that exhibit
antibacterial activity.
STATE OF THE ART
[0003] Lincomycin is a biosynthetic product that adversely affects growth of
various
microorganisms, in particular gram positive bacteria. The characteristics and
preparation of
lincomycin are disclosed in U.S. Patent 3,086,912. A variety of derivatives of
lincomycin,
which also have antimicrobial activity, have been prepared. These derivatives
include, for
example, clindamycin, which is described in U.S. Patent 3,496,163.
[0004] Lincomycin derivatives remain attractive targets for antibacterial drug
discovery.
Accordingly, lincomycin derivatives that possess antimicrobial activity are
desired as potential
antibacterial agents.
SUMMARY OF THE INVENTION
[0005] The present invention provides lincomycin derivatives that possess
antibacterial
activity.
[0006] In one of its composition aspects, this invention is directed to a
compound of
formula (I):
R3
O
R2
R9
~N HN
6 O
HO
HO '~~OH
~~iiIISR~
(I)
wherein:



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Rl is alkyl;
R2 and R3 are independently H, alkyl, hydroxy, fluoro, or cyanoalkyl or one
of R2 and R3 is NOR7 and the other is absent, or one of R2 and R3 is =CH2 and
the
other is absent, with the provisos that both R~' and R3 are not H; when one of
Ra and R3
is fluoro, the other is not hydrogen or hydroxy; and when one of R2 and R3 is
hydroxy,
the other is not fluoro, hydrogen, or hydroxy;
R6 is selected from the group consisting of H, alkyl, hydroxyalkyl,
-C(O)O-alkylene-cycloalkyl, -C(O)O-alkylene-substituted cycloalkyl, -C(O)O-
alkyl,
-C(O)O-substituted alkyl, -C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-
heteroaryl,
-C(O)O-substituted heteroaryl, -[C(O)O]p-alkylene-heterocycle, -[C(O)O]p
alkylene-
substituted heterocycle, wherein p is 0 or 1 with the proviso that -C(O)O-
substituted
alkyl does not include the following:
0
0
0
a
0
0
R' is H or alkyl;
R9, which can be singly or multiply substituted in the ring on the same or
different carbons, is independently selected from the group consisting of
hydrogen,
alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH2)n OH, -(CHa)"-NR4R5, -alkylene-Ra where Ra is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and RS are H or alkyl;
and
m is 1 or 2; and
prodrugs, tautomers or pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula I has a minimum inhibition
concentration of
32 ~.g/mL or less against at least one of the organisms selected from the
group consisting of
Streptococcus p~eumoniae, Staphylococcus aureus, Staphylococcus epide~midis,
E~rterococcus
2



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
faecalis, Enterococcus faecium, Haemophilus influenzae, Moraxella catarrhalis,
Escherichia
coli, Bacteroides fragilis, Bacteroides thetaiotaomicron, and Clostridium
diffzcile.
[0007] In a preferred embodiment, this invention provides compounds of formula
(II)
R9-
~~uISR~
(II)
wherein:
R1 is alkyl;
R2 and R3 are independently H, alkyl, or cyanoalkyl, with the proviso that
both R2 and R3 are not H;
R6 is H, alkyl, or hydroxyalkyl;
R9, which can be singly or multiply substituted in the ring on the same or
different carbons, is independently selected from the group consisting of
hydrogen,
alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CHa)n OH, -(CHa)"-NR4R$, -alkylene-Ra where Ra is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and RS are H or alkyl;
and
m is 1 or 2; and
prodrugs and pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula II has a minimum inhibition
concentration of
32 ~,g/mL or less against at least one of the organisms selected from the
group consisting of
Streptococcus pheumoniae, Staphylococcus aureus, Staphylococcus epidermidis,
Enterococcus
faecalis, E~cterococcus faecium, Haemophilus influenzae, Moraxella
catarrhalis, Escherichia
coli, Bacteroides fragilis, Bacteroides thetaiotaomicrou, and Clostridium
difficile.
(0008] In a particularly preferred embodiment, this invention provides
compounds of
formula (III):
3



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
R'
~~~SR~
wherein:
Rl is alkyl;
R2 and R3 are fluoro;
R6 is H, alkyl, or hydroxyalkyl;
R9, which can be singly or multiply substituted in the ring on the same or
different carbons, is independently selected from the group consisting of
hydrogen,
alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CH~)n OH, -(CH2)"NR4R5, -alkylene-Ra where Ra is selected from
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and RS are H or alkyl;
and
m is 1 or 2; and
prodrugs and pharmaceutically acceptable salts thereof,
with the proviso that the compound of formula III has a minimum inhibition
concentration of
32 ~g/mL or less against at least one of the organisms selected from the group
consisting of
Streptococcus p~ceumohiae, Staphylococcus aureus, Staphylococcus epidermidis,
Enterococcus
faecalis, Ehte~ocoecus faecium, Haemophilus influenzae, Moraxella cata~rhalis,
Escherichia
coli, Bacte~oides ft~agilis, Bacte~oides thetaiotaomicron, ahd Clostridium
difficile.
In another preferred embodiment, this invention is directed to a compound of
formula (IV):
4



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
R3
O
Rs
Rz
~N HN
6 O
HO
HO ~~OH
wherein:
niIISR~
(IV)
Rl is alkyl;
R2 and R3 are independently H, or alkyl, hydroxy, fluoro, or cyanoalkyl or
one of R2 and R3 is NOR7 and the other is absent, or one of Ra and R3 is =CHI
and
the other is absent, with the provisos that both Ra and R3 are not H; when one
of R2
and R3 is fluoro, the other is not hydrogen or hydroxy; and when one of R~ and
R3 is
hydroxy, the other is not fluoro, hydrogen, or hydroxy;
R6 is selected from the group consisting of -C(O)O-alkylene-cycloalkyl,
-C(O)O-alkylene-substituted cycloalkyl, -C(O)O-alkyl, -C(O)O-substituted
alkyl,
-C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-heteroaryl, -C(O)O-substituted
heteroaryl, -[C(O)O]p alkylene-heterocycle, -[C(O)O]p alkylene-substituted
heterocycle, wherein p is 0 or 1 with the proviso that -C(O)O-substituted
alkyl does
not include the following:
0
0
0 0
0
0
0
R' is H or alkyl;
R9, which can be singly or multiply substituted in the ring on the same or
different carbons, is independently selected from the group consisting of
hydrogen,
alkyl, substituted alkyl, alkoxyalkoxy, cycloalkyl, substituted cycloalkyl,
alkoxyalkoxy, substituted oxygen, substituted nitrogen, halogen, phenyl,
substituted
phenyl, -(CHa)n OH, -(CH2)"-NR4R5, -alkylene-Ra where Ra is selected from



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
monofluorophenyl and monochlorophenyl, and branched chain isomers thereof
wherein n is an integer of from 1 to 8 inclusive and R4 and RS are H or alkyl;
and
m is 1 or 2; and
prodrugs, tautomers or pharmaceutically acceptable salts thereof;
with the proviso that the compound of formula I has a minimum inhibition
concentration of
32 ~,g/mL or less against at least one of the organisms selected from the
group consisting of
Streptococcus pneumohiae, Staphylococcus aureus, Staphylococcus epidermidis,
Ehterococcus
faecalis, Enteroc~ccus faecium, Haemophilus ihfluenzae, Moraxella catarrhalis,
Escherichia
coli, Bacteroides fragilis, Bacteroides thetaiotaomicron, and Clostridium
difficile.
[0009] Lincomycin derivatives within the scope of this invention include those
set forth in
Table I as follows:
R''
Ex. R R !R R6 R~~~ m


1 Methyl H/methyl H ethyl 2


2 Methyl H/methyl methyl ropyl 1


3 Methyl H/cyanomethyl methyl propyl 1


4 Methyl hydroxy/methylH ethyl 2


Methyl H/hydroxyiminomethyl pro yl 1


6 Methyl H/methoxyiminomethyl pro yl 1


7 Methyl H/methyl H butyl 2


8 Methyl H/methyl H pentyl 1


9 Methyl H/methyl H isopentyl 1


Methyl H/methyl H pentyl 1


11 Methyl fluoro/fluoro methyl ropyl 1


12 Methyl fluoro/fluoro H pentyl 1


Includes R and/or S isomers as either individual isomres or as a mixture
6



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Ex. R R /R R Ry* m


13 Methyl H/methyl H 2-(4-fluorophenyl)ethyl1


14 Methyl H/methyl H 3,3-difluoro ro 1
yl


15 Methyl H/methyl H 2-(4-chloro henyl)ethyl1


16 Methyl H/methyl H 2,2-difluoro entyl 1


17 Methyl Hlmethyl H propyl 2


18 Methyl H/methyl 2-hydroxyethyl pentyl 1


19 Methyl H/methyl 2-methyl-2- pentyl 1
hydroxyethyl


20 Methyl H/methyl 2-methyl-2- pentyl 1
hydroxyethyl


21 Methyl H/methyl 3-hydroxypro n- entyl 1
yl


22 Methyl H/methyl 2-hydroxyethyl isopentyl 1


23 Methyl H/methyl 2-hydroxyethyl 3,3-difluoro ropyl 1


24 Methyl fluoro/fluoro 2-hydroxyethyl entyl 1


25 methyl H/methyl H pentyl 2


26 methyl H/methyl H methoxy 2


27 methyl H/methyl H 1-ethylpropyl 2


28 methyl H/methyl H iso-propyl 2


29 methyl H/methyl H butyl 2


30 methyl H/methyl H cyclohexyl 2


31 methyl H/methyl 2-hydroxyethyl ethyl 2


32 methyl H/methyl 2-hydroxyethyl pentyl 2


33 methyl H/methyl 2-hydroxyethyl propyl 2


34 methyl H/methyl fluorenylmethylenepropyl 2
oxy-carbonyl


35 methyl H/methyl ethoxycarbonyl propyl 2


36 methyl H/methyl phenoxycarbonylpropyl , 2


37 methyl H/methyl H 4-difluoropentyl 1


38 methyl H/methyl H 3-difluorobutyl 1


39 methyl H/methyl 2-hydroxyethyl 3-difluoropentyl 1


40 methyl R2/R3 = vinyl H butyl 1


41 methyl H/methyl H 3-difluoropropyl 2


42 methyl H/methyl H 3-difluorobutyl 2


43 methyl H/methyl H 5-difluoropentyl 2
,


44 methyl H/methyl H 5-fluoropentyl 2


45 methyl H/methyl H 4-fluorobutyl 2





CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Ex. R R /R R R m


46 methyl H/methyl H 3-hydroxy-3-ethylpentyl2


47 methyl H/methyl H butoxy 2


48 methyl H/methyl H pentoxy 2


49 methyl H/methyl H 4-fluorobutoxy 2


51 methyl H/methyl ethyl ethyl 2


52 methyl H/methyl H 3-fluoropropoxy 2


53 methyl H/methyl H 3-trifluoropropoxy 2


54 methyl H/methyl H isobutyl 2


55 methyl fluoro/fluoro H propyl 2


56 methyl H/methyl H fluorolpropyl 1


57 methyl H/methyl H fluoro/butyl 1


58 Methyl H/hydroxy H fluoro/pro yl 1


59 methyl H/methyl H 2-methoxyethoxy


60 methyl H/methyl H butyl 1


61 methyl H/methyl H 4-difluoropentyl 2


62 methyl H/methyl H 3-fluoropropyl 2


63 methyl Hlmethyl H fluoro/propyl 2


64 methyl H/methyl H 2-fluoroethoxy 2


65 methyl H/methyl H 3-difluoropentyl 1


(0010] As used below, these compounds are named based on acetamide derivatives
but,
alternatively, these compounds could have been named based on 1-thio-L-threo-a-
D-galacto-
octopyranoside derivatives. Specific compounds within the scope of this
invention include the
following compounds:
1-(4-ethylpiperid-6-yl)-N-{ 1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(-4-n-propyl-N-methylpyrrolidin-2-yl)-N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide ;
1-(-4-n-propyl-N-methylpyrrolidin-2-yl)-N-{ 1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methyl-3-cyanoprop-1-yl}acetamide;
8



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
1-(-4-ethylpiperidyl)-N-{ 1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-hydroxy-2-methylprop-1-yl}acetamide;
1-(-4-n-propyl-N-methylpyrrolidin-2-yl)-N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-hydroxyiminoprop-1-yl} acetamide;
1-(-4-n-propyl-N-methylpyrrolidin-2-yl) N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methoxyiminoprop-1-yl } acetamide;
1-(-3-n-butylpiperid-6-yl)-N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide ;
1-(4-n-pentylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4- (3-methylbut-1-yl)pyrrolidin-2-yl]-N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(-4-n-pentylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(-4-n-propyl-N-methylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2,2-difluoroprop-1-yl}acetamide ;
1-(-4-n-pentylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2,2-difluoroprop-1-yl}acetamide ;
1-(-4-(3-p-fluorophenyl)prop-1-ylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-
6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-(3,3-difluoroprop-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(-4-(3-p-chlorophenyl)prop-1-ylpyrrolidin-2-yl)-N-{ 1-[3,4,5-trihydroxy-
6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-(2,2-difluoropent-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide ;
1-(-4-n-propylpiperid-6-yl)-N-{1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-[4-n-pentyl-N-(2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-n-pentyl-N-(2-(R)-methyl-2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{ 1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide;



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
1-[4-n-pentyl-N-(2-(S)-methyl-2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{ 1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide;
1-(-4-n-pentyl-N-(3-hydroxyprop-1-yl)pyrrolidin-2-yl) N-{1-[3,4,5-
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4- (3-methylbut-1-yl)-N-(2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide;
1-[4- (3,3-difluoroprop-1-yl)-N-(2-hydroxyeth-1-yl)pyrrolidin-2-yl] N-{1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide;
1-[4-n-pentyl-N-(2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2,~-difluoroprop-1-
yl}acetamide;
1-(4-n-pentylpiperid-6-yl)-N-{ [3, 4, 5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-[4-(1-ethylprop-1-yl)piperid-6-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-iso-propylpiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-n-butylpiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-cyclohexylpiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-( 4-ethyl-N-hydroxyethyl-piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-( 4-n-pentyl-N-hydroxyethyl-piperid-6-yl)=N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-( 4-n-propyl-N-hydroxyethyl-piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-[4-n-propyl-N-(F-moc)-piperid-6-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl } acetamide;
1-[4-n-propyl-N-(carboxylic acid ethyl ester)-piperid-6-yl]-N-{ 1-[3,4,5-
trihydroxy-6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-n-propyl-N-(carboxylic acid phenyl ester)-piperid-6-yl]-N-{1-[3,4,5-
trihydroxy-6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl } acetamide;



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
1-[4- (4,4-difluoropent-1-yl) pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-(3,3-difluorobut-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-(3,3-difluoropent-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-(3,3-difluoropent-1-yl)-N-(2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N-{ 1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide;
1-(4-(2,2-difluoroeth-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(4-(3,3-difluoroprop-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-(5,5-difluoropent-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(4-(5-fluoropent-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-(4-fluorobut-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-(3-ethyl-3-hydroxypent-1-yl)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-butoxypiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(4-pentoxypiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
1-(4-(4-fluorobutoxy)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-n-butylprop-1-yl)pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methyl-allyl } acetamide;
1-( 4-ethyl-N-ethyl-piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-(3-fluoropropoxy)piperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
11



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
1-(4-(3,3,3-trifluoropropoxy)piperid-6-yl) N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl } acetamide;
1-(4-iso-butylpiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-(4-n-propylpiperid-6-yl)-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2,2-difluoro-prop-1-yl} acetamide;
1-[4-n-propyl-4-fluoro-pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide;
1-[4-n-butyl-4-fluoro-pyrrolidin-2-yl]-N-{ 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide;
4-Fluoro-4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxy-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide;
4-Fluoro-4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxy-1-(3,4,5-
trihydroxy-6-methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amid; and
and prodrugs, tautomers and pharmaceutically acceptable salts thereof.
[0011] The compounds, tautomers, prodrugs and pharmaceutically acceptable
salts thereof,
as defined herein, may have activity against bacteria, protozoa, fungi, and
parasites.
[0012] In another aspect, this invention provides pharmaceutical compositions
comprising a
pharmaceutically acceptable carrier and a therapeutically effective amount of
the compounds
defined herein. The pharmaceutical compositions of the present invention may
further comprise
one or more additional antibacterial agents, one of which may be active
against gram negative
bacteria. One of which may also be active against gram positive bacteria.
[0013] In one of its method aspects, this invention is directed to a method
for the treatment
of a microbial infection in a mammal comprising administering to the mammal a
therapeutically
effective amount of a compound of this invention. The compound of this
invention may be
administered to the mammal orally, parenterally, transdermally, topically,
rectally, or
intranasally in a pharmaceutical composition.
[0014] In another of its method aspects, this invention is directed to a
method for the
treatment of a microbial infection in a mammal comprising administering to the
mammal a
pharmaceutical composition comprising a therapeutically effective amount of a
compound of
this invention. The pharmaceutical compositions of the present invention may
further comprise
one or more additional antibacterial agents, one of which may be active
against gram negative
bacteria. One of which may also be active against gram positive bacteria. The
pharmaceutical
12



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
composition may be administered to the mammal orally, parenterally,
transdermally, topically,
rectally, or intranasally.
[0015] In a preferred embodiment, the microbial infection being treated is a
gram positive
infection. In an additional embodiment, the infection may be a gram negative
infection. In a
further embodiment, the infection may be a mycobacteria infection, a
mycoplasma infection, or
a chlamydia infection.
[0016] In yet another aspect, the present invention provides novel
intermediates and
processes for preparing compounds of formula (I), (II), and (III).
DETAILED DESCRIPTION OF THE 1NVENTION
[0017] As described above, this invention relates to lincomycin derivatives
that exhibit
antibacterial activity, in particular gram positive antibacterial activity.
However, prior to
describing this invention in further detail, the following terms will first be
defined.
Definitions
[0018] Unless otherwise stated, the following terms used in the specification
and claims
have the meanings given below.
[0019] "AcyY' means the group -C(O)R' wherein R' is alkyl, alkenyl, alkynyl,
aryl,
substituted aryl, heteroaryl, or substituted heteroaryl.
[0020] "Acyloxy" means the group -C(O)OR', wherein R' is alkyl, alkenyl,
alkynyl, aryl,
substituted aryl, heteroaryl, or substituted heteroaryl.
[0021] "Alkenyl" means a linear unsaturated monovalent hydrocarbon radical of
two to
eight carbon atoms or a branched monovalent hydrocarbon radical of three to
eight carbon atoms
containing at least one double bond, (-C=C-). Examples of alkenyl groups
include, but are not
limited to, allyl, vinyl, 2-butenyl, and the like.
[0022] "Alkoxy" refers to the group "alkyl-O" which includes, by way of
example,
methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-
pentoxy, n-
hexoxy, 1,2-dimethylbutoxy, and the like.
[0023] "Alkoxyalkoxy" refers to the group alkyl-O-alkylene-O-, wherein alkyl
is as defined
herein.
[0024] "Alkyl" means a linear saturated monovalent hydrocarbon radical of one
to eight
carbon atoms or a branched saturated monovalent hydrocarbon radical of three
to eight carbon
atoms. Examples of alkyl groups include, but are not limited to, groups such
as methyl, ethyl, n-
propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-phenyl, and the
like.
13



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0025] "Alkylene" means a linear divalent hydrocarbon radical of one to eight
carbon atoms
or a branched divalent hydrocarbon group of three to eight carbon atoms.
Examples of alkylene
groups include, but are not limited ~to, methylene, ethylene, 2-
methylpropylene, and the like.
[0026] "Alkylthio" refers to the group "alkyl-S-" which includes, by way of
example,
methylthio, butylthio, and the like.
[0027] "Alkynyl" means a linear monovalent hydrocarbon radical of two to eight
carbon
atoms or a branched monovalent hydrocarbon radical of three to eight carbon
atoms containing
at least one triple bond, (-C=C-). Examples of alkynyl groups include, but are
not limited to,
ethynyl, propynyl, 2-butynyl, and the like.
[0028] "Amino" or "substituted nitrogen" refers to the group "-NRaRb" wherein
Ra and Rb
are independently H, alkyl, haloalkyl, alkenyl, cycloalkyl, substituted
cycloalkyl, aryl,
substituted aryl, heteroaryl, or substituted heteroaryl.
[0029] "Aminocarboxyalkyl" means a group "-R~C(O)NRaRb" where R~ is an
alkylene, as
defined above, and Ra and Rb are as defined above.
[0030] "Aryl" means a monovalent monocyclic or bicyclic aromatic carbocyclic
group of six
to fourteen ring atoms. Examples include, but are not limited to, phenyl,
naphthyl, and anthryl.
The aryl ring may be optionally fused to a 5-, 6-, or 7-membered monocyclic
non-aromatic ring
optionally containing 1 or 2 heteroatoms independently selected from oxygen,
nitrogen, or
sulfur, the remaining ring atoms being C where one or two C atoms are
optionally replaced by a
carbonyl. Representative aryl groups with fused rings include, but are not
limited to, 2,5-
dihydro-benzo[b]oxepinyl, 2,3-dihydrobenzo[1,4]dioxanyl, chromanyl,
isochromanyl, 2,3-
dihydrobenzofuranyl, 1,3-dihydroisobenzofuranyl, benzo[1,3]dioxolyl, 1,2,3,4-
tetrahydroisoquinolinyl, 1,2,3,4-tetrahydroquinolinyl, 2,3-dihydro-1H-indolyl,
2,3dihydro-1H-
isoindolyl, benzimidazole-2-onyl, 2-H-benzoxazol-2-onyl, and the like.
[0031] "Carbonyl" means the group "C(O)."
[0032] "Carboxy" means the group "C(0)O."
[0033] "Cyanoalkyl" refers to an alkyl, wherein alkyl is as defined above,
substituted with
one or more cyano (-CN) groups provided that if two cyano groups are present
they are not both
on the same carbon atom. Examples of cyanoalkyl groups include, for example,
cyanomethyl,
2-cyanoethyl, 2-cyanopropyl, and the like.
[0034] "Cycloalkyl" refers to cyclic alkyl groups of from 3 to 20 carbon atoms
having a
single or multiple cyclic rings including, by way of example, cyclopropyl,
cyclobutyl,
cyclopentyl, cyclooctyl, adamantanyl, and the like. Cycloalkyl groups of the
present invention
14



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
also include fused multicyclic rings wherein one or more of the rings within
the multicyclic ring
system are aromatic, as long as the point of attachment to the core or
backbone of the structure is
on the non-aromatic ring, e.g., fluorenyl.
(0035] "Cycloalkylalkyl" means a group -R~Rd where R~, is an alkylene group
and Rd is a
cycloalkyl group, as defined above. Examples include, but are not limited to,
cyclopropylmethylene, cyclohexylethylene, and the like.
[0036] "Halo" or "Halogen" means fluoro, chloro, bromo, or iodo.
[0037] "Haloalkyl" means an alkyl, wherein alkyl is as defined above,
substituted with one
or more, preferably one to 6, of the same or different halo atoms. Examples of
haloalkyl groups
include, for example, trifluoromethyl, 3-fluoropropyl, 2,2-dichloroethyl, and
the like.
[0038] "Heteroaryl" means a monovalent monocyclic or bicyclic aromatic radical
of 5 to 10
ring atoms containing one, two, or three ring heteroatoms selected from N; O,
or S, the
remaining ring. atoms being C. Representative examples include, but are not
limited to, thienyl,
benzothienyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinolinyl,
quinoxalinyl, imidazolyl, .
furanyl, benzofuranyl, thiazolyl, isoxazolyl, benzisoxazolyl, benzimidazolyl,
triazolyl, pyrazolyl,
pyrrolyl, indolyl, 2-pyridonyl, 4-pyridonyl, N-alkyl-2-pyridonyl, pyrazinonyl,
pyridazinonyl,
pyrimidinonyl, oxazolonyl, and the like.
[0039] "Heterocycle" or "heterocyclic" refers to a saturated or unsaturated
group having a
single ring or multiple condensed rings, from 1 to 10 carbon atoms and from 1
to 4 heteroatoms
selected from the group consisting of nitrogen, sulfur, or oxygen within the
ring, wherein, in
fused ring systems one or more of the rings can be aryl or heteroaryl as
defined herein.
Examples of heterocycles and heteroaryls include, but are not limited to,
azetidine, pyrrole,
imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine,
isoindole, indole,
dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline,
phthalazine,
naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole,
carboline,
phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole,
phenoxazine,
phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline,
phthalimide, 1,2,3,4-
tetrahydro-isoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazole,
thiazolidine, thiophene,
benzo[b]thiophene, morpholinyl, thiomorpholinyl (also referred to as
thiamorpholinyl),
piperidinyl, pyrrolidine, tetrahydrofuranyl, and the like.
[0040] Heterocycles may be optionally substituted with from one to three
substituents
selected from the group consisting of alkyl, alkenyl, alkynyl, halo, alkoxy,
acyloxy, amino,
hydroxyl, carboxy, cyano, oxo, vitro, and alkylthio as these terms are defined
herein.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0041] "Hydroxy" or "hydroxyl" means the group -OH.
[0042] "Hydroxyalkyl" refers to an alkyl, wherein alkyl is as defined above
substituted with
one or more -OH groups provided that if two hydroxy groups are present they
are not both on
the same carbon atom. Examples of hydroxyalkyl groups include, for example,
hydroxymethyl,
2-hydroxyethyl, 2-hydroxypropyl, and the like.
[0043] "Mammal" refers to all mammals including humans, livestock, and
companion
animals.
[0044] "Optional" or "optionally" means that the subsequently described event
or
circumstance may, but need not, occur, and that the description includes
instances where the
event or circumstance occurs and instances in which it does not. For example,
"aryl group
optionally mono- or di- substituted with an alkyl group" means that the alkyl
may but need not
be present, and the description includes situations where the aryl group is
mono- or disubstituted
with an alkyl group and situations where the aryl group is not substituted
with the alkyl group.
[0045] "Pharmaceutically acceptable carrier" means a carrier that is useful in
preparing a
pharmaceutical composition that is generally safe, non-toxic and neither
biologically nor
otherwise undesirable, and includes a carrier that is acceptable for
veterinary use as well as
human pharmaceutical use. "A pharmaceutically acceptable carrier" as used in
the specification
and claims includes both one and more than one such carrier.
[0046] "Pharmaceutically acceptable salt" of a compound means a salt that is
pharmaceutically acceptable and that possesses the desired pharmacological
activity of the
parent compound. Such salts include, but are not limited to,
(1) acid addition salts, formed with inorganic acids such as hydrochloric
acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the
like; or
formed with organic acids such as acetic acid, propionic acid, hexanoic acid,
cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic
acid,
succinic acid, malic acid, malefic acid, fumaric acid, tartaric acid, citric
acid, benzoic
acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid,
methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-
hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic
acid, 2-
naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-
methylbicyclo[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid,
4,4'methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid,
trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid,
gluconic acid,
16



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic
acid, and
the like; or
(2) salts formed when an acidic proton present in the parent compound
either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline
earth metal ion,
or an aluminum ion; or coordinates with an organic base such as ethanolamine,
diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the
like.
[0047] "Prodrugs" mean any compound which releases an active parent drug
according to a
compound of the subject invention in vivo when such prodrug is administered to
a mammalian
subject. Prodrugs of a compound of the subject invention are prepared by
modifying functional
groups present in a compound of the subject invention in such a way that the
modifications may
be cleaved in vivo to release the parent compound. Prodrugs include compounds
of the subject
invention wherein a hydroxy, sulfhydryl or amino group in the compound is
bonded to any
group that may be cleaved ih vivo to regenerate the free hydroxyl, amino, or
sulfliydryl group,
respectively. Examples of prodrugs include, but are not limited to esters
(e.g., acetate, formate,
and benzoate derivatives), carbamates (e.g., N,N-dimethylaminocarbonyl) of
hydroxy functional
groups in compounds of the subject invention, and the like. Specific examples
include
-C(O)O-alkylene-cycloalkyl, -C(O)O-alkylene-substituted cycloalkyl, -C(O)O-
alkyl,
-C(O)O-substituted alkyl, -C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-
heteroaryl,
-C(O)O-substituted heteroaryl, -[C(O)O]p alkylene-heterocycle, -[C(O)O]p
alkylene-substituted
heterocycle, wherein p is 0 or 1 with the proviso that -C(O)O-substituted
alkyl does not include
the following:
0
a
0
0
0
0
[0048] "Substituted alkyl" means an alkyl group, as defined above, in which
one or more of
the hydrogen atoms has been replaced by a halogen (i.e., Cl, Br, F, or I),
oxygen, hydroxy,
amine (primary), amine (secondary-alkyl substituted by alkyl above) amine
(tertiary-alkyl
substituted by alkyl as above), sulfur, -SH, or phenyl. Examples of
substituted alkyl groups
17



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
include, but are not limited to, 1-fluoroethyl, 1-chloroethyl, 2-fluoroethyl,
2-chloroethyl, 1-
bromopropyl, 2-iodopropyl, 1-chlorobutyl, 4-flurobutyl, and 4-chlorobutyl.
[0049] "Substituted aryl" means an aryl ring. substituted with one or more
substituents,
preferably one to three substituents selected from the group consisting of
alkyl, alkenyl, alkynyl,
halo, alkoxy, acyloxy, amino, hydroxy, carboxy, cyano, vitro, alkylthio, and
thioalkyl. The aryl
ring may be optionally fused to a 5-, 6-, or 7-membered monocyclic non-
aromatic ring
optionally containing 1 or 2 heteroatoms independently selected from oxygen,
nitrogen, or
sulfur, the remaining ring atoms being carbon where one or two carbon atoms
are optionally
replaced by a carbonyl.
[0050] "Substituted cycloalkyl" means a cycloalkyl substituted with an alkyl
group, wherein
alkyl is as defined above or a group as defined above for substituted alkyl.
Representative
examples include, but are not limited to, 2-cyclopropylethyl, 3-
cyclobutylpropyl, 4-
cyclopentylbutyl, 4-cyclohexylbutyl, and the like.
[0051] "Substituted heteroaryl" means a heteroaryl ring, wherein heteroaryl is
as defined
above, substituted with one or more substituents, preferably one to three
substituents selected
from the group consisting of alkyl, alkenyl, alkynyl, halo, alkoxy, acyloxy,
amino, hydroxy,
carboxy, cyano, vitro, alkylthio, and thioalkyl, wherein said substituents are
as defined herein.
[0052] "Substituted oxygen" refers to the group "-O-R~" wherein Rd is alkyl,
haloalkyl,
alkenyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
heteroaryl, or substituted
heteroaryl, wherein said substituents are as defined herein.
[0053] "Substituted phenyl" means a phenyl ring wherein one or more of the
hydrogen
atoms has been replaced by a halogen, hydroxy, alkyl, amine (primary,
secondary, and tertiary
with the latter two alkyl substituted), -SH, and phenyl. Representative
examples include, but are
not limited to, p-bromophenyl, m-iodophenyl, o-chlorophenyl, p-ethylphenyl, m-
propylphenyl,
o-methylphenyl, and p-octylphenyl.
(0054] "Thioalkyl" refers to an alkyl, wherein alkyl is as defined above,
substituted with one
or more -SH groups provided that if two hydroxy groups are present they are
not both on the
same carbon atom. Examples of thioalkyl groups include, for example,
thiomethyl, 2-thioethyl,
2-thiopropyl, and the like.
[0055] "Therapeutically effective amount" means the amount of a compound or
composition
that, when administered to a mammal for treating a disease, is sufficient to
effect such treatment
for the disease. The "therapeutically effective amount" will vary depending on
the compound or
composition, the disease and its severity and the age, weight, etc., of the
mammal to be treated.
18



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0056] "Treating" or "treatment" of a disease includes:
(1) preventing the disease, i.e. causing the clinical symptoms of the disease
not to develop in a mammal that may be exposed to or predisposed to the
disease but
does not yet experience or display symptoms of the disease,
(2) inhibiting the disease, i.e., arresting or reducing the development of the
disease or its clinical symptoms, or
(3) relieving the disease, i.e., causing regression of the disease or its
clinical symptoms.
"Tautomer" refers to an isomer in which migration of a hydrogen atom
results in two or more structures.
[0057] The compounds of the present invention are generally named according to
the
IUPAC or CAS nomenclature system. Abbreviations that are well known to one of
ordinary
skill in the art may be used (e.g. "Ph" for phenyl, "Me" for methyl, "Et" for
ethyl, '.'Bn" for
benzyl, "h" for hour and "rt" for room temperature).
General Synthetic Schemes
[0058] Compounds of this invention can be made by the methods depicted in the
reaction
schemes shown below.
[0059] The starting materials and reagents used in preparing these compounds
are either
available from commercial suppliers such as Toronto Research Chemicals (North
York, ON
Canada), Aldrich Chemical Co. (Milwaukee, Wisconsin, USA), Bachem (Torrance,
California,
USA), Emka-Chemie, or Sigma (St. Louis, Missouri, USA) or are prepared by
methods known
to those skilled in the art following procedures set forth in references such
as Fieser and Fieser's
Reagents for Organic Synthesis, Volumes 1-15 (John Wiley and Sons, 1991),
Rodd's Chemistry
of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science
Publishers, 1989),
Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced
Organic
Chemistry, (John Wiley and Sons, 4th Edition), and Larock's Comprehensive
Organic
Transformations (VCH Publishers Inc., 1989). These schemes are merely
illustrative of some
methods by which the compounds of this invention can be synthesized, and
various
modifications to these schemes can be made and will be suggested to one
skilled in the art
having referred to this disclosure.
[0060] As it will be apparent to those skilled in the art, conventional
protecting groups may
be necessary to prevent certain functional groups from undergoing undesired
reactions. Suitable
19



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
protecting groups for various functional groups, as well as suitable
conditions for protecting and
deprotecting particular function groups are weh known in the art. For example,
numerous
protecting groups are described in T.W. Greene and G.M. Wuts, P~otecti~cg
Groups in Organic
Synthesis, Second Edition, Wiley, New York, 1991, and references cited
therein.
[0061] The starting materials and the intermediates of the reaction may be
isolated and
purified if desired using conventional techniques, including but not limited
to filtration,
distillation, crystallization, chromatography, and the like. Such materials
may be characterized
using conventional means, including physical constants and spectral data.
[0062] The compounds of this invention will typically contain one or more
chiral centers.
Accordingly, if desired, such compounds can be prepared or. isolated as pure
stereoisomers. All ..
such stereoisomers (and enriched mixtures) are included within the scope of
this invention,
unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be
prepared using,
for example, optically active starting materials or stereoselective reagents
well-known in the art.
Alternatively, racemic mixtures of such compounds can be separated using, for
example, chiral
column chromatography, chiral resolving agents, and the like.
Preparation of Compounds of formula (I)
[0063] In general, to prepare the compounds of formula (I) of the present
invention, an
appropriately 7-substititued lincosamine intermediate and an appropriately
substituted
pyrrolidinyl or piperidyl carboxylic acid are condensed under reactive
conditions, preferably. in
an inert organic solvent, in the presence of a coupling reagent and an organic
base. This reaction
can be performed with any number of known coupling reagents, such as O-(7-
azabenzotriazol-1-
yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU), 1-
hydroxybenzotriazole
hydrate (HOBT) with carbodiimides, isobutyl chloroformate, diphenylphosphoryl
azide (DPPA),
and the like. Suitable organic bases include diisopropylethylamine (DIEA),
triethylamine
(TEA), pyridine, N-methyl morpholine, and the like. Suitable inert organic
solvents which can
be used include, for example, N,N-dimethylformamide, acetonitrile,
dichloromethane, and the
like. This reaction is typically conducted using an excess of carboxylic acid
to lincosamine at
temperatures in the range of about 0°C to about 50°C. The
reaction is continued until
completion, which typically occurs in from about 2 to 12 h.
[0064] Appropriately 7-substititued lincosamine intermediates, as defined in
the present
invention (i. e., R2/R3), are synthesized by methods well known to those of
skill in the art from
methyl 6-amino-6,8-dideoxy-1-thin-erythro-a-D-galacto-octopyranoside, which
can be prepared



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
as described by Hoeksema, et al., Journal of the American Chemical
Society,1967, 89 2448-
2452. Illustrative syntheses for 7-substituted lincosamine intermediates are
shown below in
Schemes 1-5.
[0065] Appropriately substituted pyrrolidinyl or piperidyl carboxylic acid
intermediates, as
defined in the present invention (i.e., R9), are also synthesized by methods
well known to those
of skill in the art from prolines and pyridines. The prolines and pyridines
that can be used in the
synthesis of the carboxylic acid intermediates of the present invention
include, for example, 4-
oxoproline and 4-substituted pyridines. The prolines and pyridines used in the
synthesis are
commercially available from vendors such as Aldrich and Sigma. Alternatively,
these prolines
and pyridines can be prepared by methods well known in the art. Illustrative
syntheses for
appropriately substituted pyrrolidinyl or piperidyl carboxylic acid
intermediates are shown
below in Schemes 6-10.
[0066] Scheme 1 below illustrates a general synthesis of a lincosamine
intermediate lc
wherein P is an N-protecting group, preferably either Cbz or Boc, and Rl is as
defined for
formula (I).
HzNn" OH
PHNn,. _O
PHNu~. OTMS
O a, b 0 c O
HO ~~~~SR~ TMSO w~~SR~ ' TMSO ~~~SR1
HO ~OH TMSOYOTMS TMSO ~OTMS
1a 1b 1c
Scheme 1. General synthesis of lincosamine intermediate lc.
(a) N-Protection (Boc, Cbz); (b) O-silyl protection (TMS); (c) Swern
oxidation.
[0067] As shown in Scheme 1, methyl 6-amino-6,8-dideoxy-1-thio-erythro-a-D-
galacto-
octopyranoside,1a, is prepared as described by Hoeksema, et al., Journal of
the American
Chemical Society,1967, 89 2448-2452. The amino functional group and the
hydroxy functional
groups of the product la are then protected with suitable protecting groups.
Suitable N-
protecting groups can be formed by the addition of di-t-butyldicarbonate, N-
(benzyloxycarbonyloxy) succinimide, and the like. The hydroxy groups can be
protected as silyl
ethers. The hydroxyl group can be converted to trimethylsilyl (TMS) ethers by
reaction with N,
O-bis-(trimethylsilyl)trifluoroacetamide in the presence of an appropriate
organic base such as
triethylamine (TEA) or trimethylsilyl chloride in the presence of an organic
base such as
triethylamine. The N-protection is typically accomplished before the O-
protection.
21



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Chromatography of the crude product on silica after evaporation of the solvent
provides the
protected product lb.
[0068] The 7-O-trimethylsilyl group of lb is chemoselectively deprotected and
oxidized to
provide the 7-keto-lincosamine derivative lc. This selective transformation is
performed by
addition of the protected product lb to dimethylsulfoxide and oxalyl chloride
in an inert organic
solvent such as dichloromethane followed by an appropriate organic base such
as triethylamine.
Alternatively, the transformation may be performed by addition of lb to
dimethyl sulfoxide and
an appropriate activating agent such as trifluoroacetic anhydride in an inert
organic solvent. The
reaction is typically conducted at temperatures in the range of approximately -
70°C to 80°C.
The resulting reaction mixture is stirred at the low temperature and is then
allowed to warm to
approximately -50°C. The reaction is maintained at this second
temperature for approximately 1
h to 3 h. To the reaction mixture is added a suitable organic base, such as
TEA, pyridine, and
the like. The reaction mixture is appropriately worked up to provide the
product lc. The
general class of conditions used in the transformation of lb to lc is known in
the art as Swern
oxidation conditions.
[0069] Scheme 2 below illustrates a general synthesis of a lincosamine
intermediate 2b
wherein P is an N-protecting group, preferably either Cbz or Boc, Rl is as
defined for
formula (I), and one of Ra and R3 is hydrogen and the other is as defined for
formula (I).
Rx
PHNn,. R2
H2Nn..
a O b O
1c TMSO ~~~SR~ HO w~~SR~
_ c
TMSO OTMS HO ~OH
2a 2b
Scheme 2. General synthesis of lincosamine intermediate 2b.
(a) Wittig olefination (RaPPh3~''X , R2P0(OEt)2, base, solvent); (b) and (c)
H2/Pd, Global de-
protection
[0070] As shown in Scheme 2, a keto-lincosamine intermediate lc is reacted to
form an
alkene using the Wittig or Homer-Wadsworth-Emmons reaction. In this reaction,
a suitable
phosphonium salt or phosphonate is deprotonated using a strong base to form a
phosphorus
ylide. Suitable phosphonium salts which can be used are
alkyltriphenylphosphonium halides,
which can be prepared by the reaction of triphenylphosphine and an alkyl
halide. Suitable
phosphorous compounds include, for example, methyltriphenylphosphonium
bromide,
diethyl(cyanomethyl)phosphonate and the like. Suitable strong bases which can
be used to form
22



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
the ylide include organolithium reagents, potassium test-butoxide, and the
like. The formation
of the phosphorus ylide is typically conducted under an inert atmosphere, such
as Na, in an inert
organic solvent such as toluene, THF, and the like, at low temperatures.
[0071] After formation of the phosphorus ylide, the product lc is added to the
reaction. The
reaction conveniently can be performed at temperatures between -40°C
and room temperature
and is stirred until completion, typically 1 to 4 h. The resulting organic
solution is worked-up
and chromatography of the crude product on silica provides the alkene product
2a.
[0072] Optionally, the product of 2a may be purified using conventional
techniques, such as
chromatography and said purified product may be used in the subsequent
coupling reaction to
yield vinyl lincosamine derivatives of the present invention.
[0073] The product 2a is then hydrogenated to provide the saturated product
2b. The
hydrogenation is typically performed in a polar organic solvent such as
methanol, ethanol, and
the like, using 10% palladium on carbon in a Parr bottle. The bottle is
purged, and charged with
Ha to approximately 50 to 70 psi and shaken until completion, typically
approximately 12 to
24 h. The resulting reaction mixture is filtered, e.g., through celite, and
rinsed with a polar
organic solvent such as methanol. The organic solution is worked up by
transferring to a resin
funnel containing dry, washed Dowex SOw-400x H+ form and shaken. After washing
the resin
with methanol and water, the product 2b is eluted from the resin by washing
with 5% TEA in
MeOH. The product can also be purified by silica gel column chromatography.
[0074] Scheme 3 illustrates a general synthesis of a lincosamine intermediate
3b wherein P
is an N-protecting group, preferably either Cbz or Boc, Rl is as defined for
formula (I), and one
of Ra or R3 is alkyl and the other is -OH.
OH OH
R2
HzNu..
PHN~". Rz
a O b O
1c TMSO w~~SR~ HO ~"~SR~
TMSO ~OTMS HO OH
3a 3b
Scheme 3. General synthesis of lincosamine intermediate 3b.
(a) R2M (carbon nucleophile); (b) (i) TMS de-protection (I~ or F-) and (ii) N-
deprotection
[0075] As shown in Scheme 3, suitable carbon nucleophiles add to 7-
ketolincosamine
intermediate lc in suitable inert organic solvents to provide a 7-hydroxy
lincosamine
intermediate 3b. Suitable carbon nucleophiles include methylmagnesium
chloride, diethyl zinc,
sodium acetylide and the like and suitable inert organic solvents which can be
used include
23



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
THF, diethyl ether, toluene, and the like. The reaction is typically conducted
at reduced
temperatures, approximately at 0°C, for about 3 to 5 h. The reaction is
then quenched with a
saturated aqueous acidic solution, such as saturated aqueous NH4C1/HZO. The
quenched
mixture is then worked up and can be purified by chromatography to provide the
product 3b.
[0076] Scheme 4 below illustrates a general synthesis of a lincosamine
intermediate 4b
wherein P is a N-protecting group, preferably Boc, Ri is as defined for
formula (I), and R2/R3 is
an oxime (--NOR'), wherein R7 is as defined for formula (I).
NOR7
H2Nn..
PHN~~~. NORM
a O b O
~~~~SR~ HO ~~~~SR~
1c TMSO
TMSO ~OTMS HO ~OH
4a 4b
Scheme 4. General synthesis of 7-oxime-lincosamines 4b.
[0077] As shown in Scheme 4, the lincosamine intermediate lc is converted to
the oxime by
stirring in the presence of a suitable reagent such as O-
trimethylsilylhydroxylamine, O-
alkylhydroxylamine hydrochloride (for example, D-methylhydroxylamine
hydrochloride), and
the like. The reaction is typically conducted in a polar organic solvent such
as methanol. The
reaction conveniently can be conducted at rt in approximately 8 to 24 h. The
solvent is removed
to provide the N-protected product 4a.
[0078] Removal of the protecting group can be carried out with acids, such as
trifluoroacetic
acid (TFA), hydrochloric acid, p-toluenesulfonic acid, and the like, in an
inert organic solvent
such as dichloromethane, dichloroethane, dioxane, THF, and the like. The
removal is typically
conducted at low temperatures, e.g., 0°C, and then gradually allowed to
warm to room
temperature to provide the product 4b.
[0079] Scheme 5 below illustrates a general synthesis of a lincosamine
intermediate Sb
wherein R2 and R3 are both fluorine, P is an N-protecting group, preferably
Cbz or Boc, and Rl
is as defined for formula (I).
F
HZNu.. F
PHN~~~. O
a, b O c.d p
1c Ac0 ~~~~SR~ Ac0 ~~~~SR~
Ac0 ~OAc Ac0 ~OAc
5a 5b
Scheme 5. General synthesis of 7-deoxy-7,7-difluorolincosamines Sb.
24



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
(a) F ; (b) AcaO, pyridine, DMAP; (c) DAST; (d) TFA
[0080] As shown in Scheme 5, the lincosamine intermediate lc is contacted with
a suitable
fluoride in an inert organic solvent. Suitable fluorides which can be used
include
tetrabutylammonium fluoride, Amberlite resin A-26 F form, HF~pyridine and the
like. Suitable
inert organic solvents include THF, acetonitrile, dichloromethane, dioxane,
and the like. The
reaction conveniently can be conducted at rt in about 1 to 2 h. The product
(not shown) can be
purified on a silica gel column.
[0081] The O-protecting groups on the product obtained from the column are
converted by
contact with acetic anhydride and dimethylaminopyridine (DMAP) in a suitable
mixture of an
inert organic solvent and an organic base, such as, for. example,
dichloromethane and pyridine.
The reaction conveniently can be conducted at rt in approximately 6 to 12 h.
The product can be
purified on silica gel column to provide product Sa.
[0082] The product Sa is contacted with a suitable fluorinating reagent and
then the N-
protecting group is removed to provide the product Sb. Suitable fluorinating
reagents which can
be used include, for example, dimethylaminosulfurtrifluoride,
[bis(2-methoxyethyl)amino]sulfurtrifluoride, and the like. The reaction is
typically conducted in
an inert organic solvent such as dichloromethane, ethylacetate, THF, and the
like at room
temperature in approximately 6 to 12 h.
[0083] Removal of the protecting group can be carried out with acids, such as
trifluoroacetic
acid (TFA), hydrochloric acid, p-toluenesulfonic acid, and the like, in an
inert organic solvent
such as dichloromethane, dichloroethane, dioxane, THF, and the like. The
removal is typically
conducted at low temperatures, e.g., 0°C, and then gradually allowed to
warm .to room
temperature to provide the product Sb.
[0084] Scheme 6 below illustrates a general synthesis of a proline
intermediate 6c wherein
R9 is as defined for formula (I).
Ra
O\~, a. ~ c. 1
~COZH C'..COZH ~~COzH
N b.
Bcc Boc Boc
6a 6b 6c
Scheme 6. General synthesis of cislt~a~s R9-proline intermediate mixtures 6c.
(a) R9CH2Br+ph3P , NaH, DMSO; (b) Ha/Pt
[0085] As shown in Scheme 6, the product 6c is prepared as described in
Birkenmeyer, et
al., Journal of Medicinal Chemistry 1972, I5, 1255-1259. Compound 6a is
commercially



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
available from vendors such as RSP (Scientific Research Consortium, Inc.).
Alternatively, 6a
can be prepared from commercially available protected hydroxy prolines by
methods well
known in the art. See, e.g., Demange, et al., Tetrahedron Letters 1998,
39,1169-1172.
[0086] Scheme 7 below illustrates a general synthesis of traps-R9-proline
intermediates 7d,
wherein R9 is alkyl or substituted alkyl.
R9: R9;
O~COZBn a. i, ii O~-~Cp2Bn b. i, 8 [,-.COZBn
N
Boc eoc Boc
Ta 7b 7c
Rg _
.
Tc ~COZti_
N
Td Boc
Scheme 7. General synthesis of traps-alkylprolines 7d.
(a) (i) LiHMDS, THF -78°C, (ii) bromoalkene; (b) (i) LiBHEt3, THF -
78°C, (ii) BF30Et2,
Et3SiH; (c) H2 Pd/C.
[0087] As shown in Scheme 7, a protected 4-oxoproline, 7a, is enolated with a
suitable
enolation agent and then alkylated with a suitable alkylating agent in an
inert organic solvent to
provide a lactam 7b (wherein R9~ is alkenyl), as described in the literature
procedure by Zhang,
et al., J.A.C.S. 1998,120 3894-3902. Compound 7a is commercially available
from vendors
such as Bachem. Alternatively, 7a can be prepared by methods well known in the
art. Suitable
enolating agents include LiHMDS, LiN(iPr)2, and the like, and suitable
alkylating agents include
allylic and benzylic bromides, for example, 4-bromo-2-methyl-2-butene and cis-
1-bromo-2-
pentene, allylbromide, and the like. ,
[0088] The lactam 7b is reduced using a suitable reducing agent to provide a
pyrrolidine 7c,
wherein R9' is alkenyl. The reduction is preformed by a two-step sequence
involving
superhydride reduction of the lactam to the hemiaminal and the subsequent
reduction of the
hemiaminal. Suitable reducing agents which can be used include
Et3SiH/BF3~OEt2,
Et3SiH/TiCl4, and the like.
[0089] The pyrrolidine 7c is then hydrogenated to simultaneously remove the
unsaturation in
the R9' substituent and remove the benzyl protecting group from the carboxylic
acid to provide
the product 7d. The hydrogenation is typically performed in a polar organic
solvent such as
methanol, ethanol, and the like, using 10% palladium on carbon in a Parr
bottle. The bottle is
purged, and charged with H2 to approximately 50 to 70 psi and shaken until
completion,
26



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
typically approximately 5 to 24 h. The reaction mixture is filtered, e.g.,
through a celite pad, and
washed with a polar organic solvent, such as methanol. Evaporation of the
combined washings
and filtrate affords the product 7d, wherein R9 is an alkyl or substituted
alkyl.
[0090] Scheme 8 below illustrates a general synthesis of trans-Rg-proline
intermediates 8c,
wherein R9 is alkyl or substituted alkyl.
o~ Rg
7d ~~COZBn b~ ~~COZBn
N N
Boc Boc
8a 8b
R9
8b c. C ~.CpzH
N
Boc
Sc
Scheme 8. General synthesis of trans-R9-substituted prolines 8c, wherein R9 is
alkyl or
substituted alkyl.
(a) 03, DCM, -78°C, DMS; (b) P+Ph3 salt, Base; (c) Ha, Pd/C.
[0091] As shown in Scheme 8, the product 7d is ozonolized to provide the
aldehyde 8a. The
ozonolysis reaction is typically conducted in an anhydrous inert organic
solvent, such as
dichloromethane, dioxane, THF, and the like, at low temperatures, e.g., -
78°C followed by
quenching of the reaction with a reducing agent such as DMS, Ph3P.
[0092] The aldehyde, 8a, is reacted with a suitable phosphonium salt in the
presence of a
strong base in an inert organic solvent. Suitable phosphonium salts which can
be used include,
for example, fluorobenzyl phosphonium chloride, 4-chlorobenzyl phosphonium
chloride,
dibromofluoromethane and triphenylphosphine, and the like. Suitable bases
which can be used
include potassium t-butoxide, organolithium reagents, and activated zinc.
Suitable organic
solvents which can be used include toluene, THF, dimethylacetamide, and the
like. The reaction
is typically conducted in an inert atmosphere, such as under nitrogen, with
vigorous stirring.
The reaction is typically conducted at rt to approximately 110°C for 1
to 2 h. The resulting
reaction mixture is appropriately worked-up and can be purified by
chromatography to provide
8b (wherein R9' is alkenyl).
[0093] The product 8b is then hydrogenated to provide the product 8c. The
hydrogenation is
typically performed in a polar organic solvent such as methanol, ethanol, and
the like, using
10% Palladium on carbon in a Parr bottle. The bottle is purged, and charged
with H2 to
27



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
approximately 40 to 70 psi and shaken until completion, typically
approximately 4 to 24 h. The
reaction mixture is filtered, e.g., through a celite pad and washed several
times with a polar
organic solvent, such as methanol. Evaporation of the combined washings and
filtrate affords
the product 8c, wherein R9 is an alkyl or substituted alkyl.
[0094] Scheme 9 below illustrates a general synthesis of trans-R9-proline
intermediates 9d,
wherein R9 is substituted alkyl wherein X is halo.
~N~C02Bn a. -- HO ~N~O~Bn ~ O ~N~COzBn
8a Boc 9a Boc 9b Bx
X ~N~C02Bn x ~N~COZH
9c Bx 9d Boc
Scheme 9. Example synthesis of trans-halosubstituted alkyl prolines 9d.
(a) Tetraallyltin, BF3~Et20; (b) DMSO, (COCI)2, TEA; (c) DAST (d)10% PdIC, H2
[0095] As shown in Scheme 9, aldehyde, 8a, is reduced and alkylated using a
suitable
reagent in an inert organic solvent to provide a hydroxyalkenyl substituted
proline, 9a. Suitable
reagents to reduce and alkylate the aldehyde include tetraallyltin/boron
trifluoride etherate,
allylTMS/boron trifluoride etherate and suitable inert organic solvents which
can be used
include THF, dichloromethane, and the like. The reaction is typically
conducted at low
temperatures, e.g., 0°C, for approximately 1 to 2 h. To the reaction
mixture is added a solution
of a suitable fluoride salt in water, for example potassium fluoride in water,
followed by the
addition of methanol. The reaction mixture is filtered, for example, over
celite. The product can
be purified by chromatography to provide 9a.
[0096] The hydroxyalkenyl substituted proline, 9a, is oxidized to the ketone
by contact with
a suitable oxidizing agent in an inert organic solvent. Suitable oxidizing
agents include oxalyl
chloride/DMSO, Dess Martin periodinane, and the like. Suitable inert organic
solvents include
dichloromethane, and the like. The reaction is typically conducted at reduced
temperatures, e.g.,
-72°C to -50°C, for approximately 30 min to 2 h. To the reaction
mixture is added a suitable
organic base, such as triethylamine. The reaction mixture is worked up to
provide product 9b.
28



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0097] The keto-substituted product 9b is halogenated by contact with a
suitable
halogenating agent in an inert organic solvent. Suitable halogenating agents
which can be used
include diethylaminosulfur trifluoride, [bis(2-methoxyethyl)amino] sulfur
trifluoride, and the
like. Suitable inert organic solvents which can be used include
dichloromethane, ethyl acetate,
THF, and the like. The reaction is typically conducted at low temperatures in
the range of
approximately -30°C to -78°C. The reaction mixture is gradually
allowed to warm to rt and
stirred at rt until completion, typically in 6 to 12 h. The reaction mixture
is worked up and can
be purified by chromatography to provide 9c.
[0098] The product 9c is then hydrogenated to provide the product 9d. The
hydrogenation is
typically performed in a polar organic solvent such as methanol, ethanol, and
the like, using
10% palladium on carbon in a Parr bottle. The bottle is purged, and charged
with Ha to
approximately 40 to 70 psi and shaken until completion, typically
approximately 4 to 24 h. The
reaction mixture is filtered, e.g., through a celite pad and washed several
times with a polar
organic solvent, such as methanol. Evaporation of the combined washings and
filtrate affords
the product 9d.
[0099] Scheme 10 below illustrates a general synthesis, as described in
Shuman, Journal of
Organic Chemistry. 1990, 55, 741-750, of substituted pyridine carboxylic acid
intermediates
lOb, wherein R9 is as defined for formula (I).
s a R9
a~ °' ~~
N R b. N R CN N- 'COzH
O
10a 10b
Scheme 10. General synthesis of substituted pyridin-2-yl carboxylic acids lOb.
[00100] As shown in Scheme 10, an appropriately substituted pyridine, is
contacted with a
suitable oxidizing agent in an inert organic solvent. The appropriately
substituted pyridine
starting materials are commercially available from vendors such as Aldrich and
Sigma.
Alternatively, these pyridines can be prepared by methods well known in the
art. Suitable
oxidizing agents which can be used include hydrogen peroxide, MCPBA, and the
like. The
reaction is typically conducted at reflux for 6 to 12 h. The reaction mixture
is then contacted
with a suitable cyanide reagent to provide the cyano-substituted pyridine,
10a. Suitable cyanide
reagents which can be used include trimethylsilyl cyanide, HCN, and the like.
Suitable inert
organic solvents include dichloromethane, dioxane, THF, and the like. The
reaction
29



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
conveniently can be conducted at rt in approximately 6 to 12 h. The reaction
mixture is worked
up to provide the cyano-substituted pyridine,10a.
[00101] The cyano-substituted pyridine,10a, is then hydrolyzed to provide the
pyridin-2-yl
carboxylic acid lOb by contact with a suitable acid. Suitable acids for
hydrolyzing the cyano
group to the carboxylic acid include hydrochloric acid, aqueous sulfuric acid,
and the like. The
reaction is typically conducted at reflux in 6 to 12 h.
[00102] Scheme 11 below illustrates the coupling reaction of a lincosamine
intermediate,
prepared as described above in Schemes 1-5, and a pyrrolidinyl or
piperidinyl,carboxylic acid,
prepared as described above in Schemes 6-10, wherein Rl, R2, R3, R6, and R9
are as defined for
formula (I) and P1 is a suitable O-protecting group and P2 is a suitable N-
protecting group.
Rs R°
O \ O
R
HzNu.. Rz s ~ n.. Rz C ~ o.. Rz
'~ a. Pz O b. Rs O
Pt0 .,nSR~ +~ ~~COyH p~0 ..ngR~ HO ..ngR~
PLO .~OPt mBoc PLO ~OP1 HO ~OH
11a
Rs
Ra Rs~~O Rs ~~O Ra
HzNu.. Rz Rs N HN~~.. Rz N HNn.. ~Rz
O ~. O d. H O
p~0 .~nSR~ + ~ ~ p~0 ..nSR~ -----~ HO .~nSR~
y N C02H
PLO OPT P O ~OP HO ~OW
11b~- 1
Scheme 11. General coupling and deprotection methods.
[00103] As shown iri Scheme 11, an appropriately 7-substititued lincosamine
intermediate
(prepared, for example, according to any one of Schemes 1-5) and an
appropriately substituted
pyrrolidinyl or piperidyl carboxylic acid (prepared, for example, according
to:any one of
Schemes 6-10) are condensed under reactive conditions, preferably in an inert
organic solvent,
in the presence of a coupling reagent and an organic base. This reaction can
be performed with
any number of known coupling reagents, such as O-(7-azabenzotriazol-1-yl)-
N,N,N',N'tetramethyluronium hexafluorophosphate (HATU), 1-hydroxybenzotriazole
hydrate
(HOBT) with carbodiimides, diphenylphosphoryl azide (DPPA), isobutyl
chloroformate, and the
like. Suitable organic bases include diisopropylethylamine (DIEA),
triethylamine (TEA),
pyridine, N-methyl morpholine, and the like. Suitable inert organic solvents
which can be used
include, for example, N,N dimethylformamide, acetonitrile, dichloromethane,
and the like. This
reaction is typically conducted using an excess of carboxylic acid to
lincosamine at temperatures



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
in the range of about 0°C to about 50°C. The reaction is
continued until completion, which
typically occurs in from about 2 to 12 h.
[0100] Removal of the protecting groups can be carried out with acids, such as
trifluoroacetic acid (TFA), hydrochloric acid, p-toluenesulfonic acid, and the
like, in an inert
organic solvent such as dichloromethane, dichloroethane, dioxane, THF, and the
like. The
removal is typically conducted at low temperatures, e.g., 0°C, and then
gradually allowed to
warm to room temperature to provide the product.
[0101] Also as shown in Scheme 11, an appropriately 7-substititued lincosamine
intermediate (prepared, for example, according to any one of Schemes 1-5) and
an appropriately
substituted pyridin-2-yl carboxylic acid (prepared, for example, according to
Scheme 10) are
condensed under reactive conditions, preferably in an inert organic solvent,
in the presence of a
coupling reagent and an organic base, as described above.
[0102] The pyridine llb is hydrogenated to provide the piperidyl product. The
hydrogenation is typically performed in a polar organic solvent such as
methanol, ethanol, and
the like, using platinum(IV)oxide in the presence of an acid such as HCI,
acetic acid, and the
like, in a Parr bottle. The bottle- is purged, and charged with H2 to
approximately 40 to 70 psi
and shaken until completion, typically approximately 24 h. The reaction
mixture is filtered, e.g.,
through a celite pad, and washed several times with a polar organic solvent
such as methanol.
Evaporation of the combined washings and filtrate affords the piperidyl
product.
[0103] The coupling of pyridine carboxylic acids and lincosamines to pyridine
llb followed
by reduction to the piperidyl product may also be conducted as described in
Birkenmeyer, et al.,
Journal of Medicinal Chemistry 1984, 27, 216-223.
[0104] Scheme 12 below illustrates the alkylation of the nitrogen of the
pyrrolidinyl or
piperidinyl ring, wherein R6 is alkyl, hydroxyalkyl, alkylene-substituted
heterocycle, or
alkylene-heterocycle, and Rl, R2, R3, and R9 are as defined for formula (I).
Rs Rs
~_~ ~/O
O Ra r1~ R
,". RZ C mNs HN,~,. R3
H O ~ ~ a. R O
HO w~~SR~ ~ HO ~v~SRi
MO ~OH HO ~~OH
12a 12b
Scheme 12. General synthesis of 1'-N substituted lincosamines. a. alkylating
agents
31



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0105] As shown in Scheme 12, the lincosamine 12a can be N-substituted by
contact with an
alkylating agent in the presence of a suitable base to provide a product 12b.
Suitable alkylating
agents which can be used include epoxides, alkyl bromides, and the like.
Suitable bases which
can be used include potassium carbonate, cesium carbonate triethylamine, and
the like. The
alkylation reaction is typically conducted in a polar organic solvent such as
methanol or DMF.
The alkylation reaction is typically conducted at low temperatures in the
range of 0°C to -10°C
for 10 to 20 h.
[0106] Scheme 13 below illustrates the acylation of the nitrogen of the
pyrrolidinyl or
piperidinyl ring, wherein R6 is -C(O)O-alkylene-cycloalkyl, -C(O)O-alkylene-
substituted
cycloalkyl, -C(O)O-alkyl, -C(O)O-substituted alkyl, -C(O)O-aryl, -C(O)O-
substituted aryl,
-C(O)O-heteroaryl, -C(O)O-substituted heteroaryl, -[C(O)O]-alkylene-
heterocycle,
-[C(O)O]_alkylene-substituted heterocycle, and Rl, R2, R3, and R9 are as
defined for formula (I):
R9-
12a
101
IZ6-X
Scheme 13. General synthesis of 1'-N substituted lincosamines.
[0107] As shown in Scheme 13, the lincosamine 12a can be N-substituted by
contact with an
acyl chloride 101, such as R6-X, wherein X is a suitable leaving group, and is
preferably
halogen, even more preferably chloride in the presence of a suitable base to
provide a product
102. Examples of compound 101, include bromofluorenyl, Cl-C(O)O-alkyl, Cl-
C(O)O-aryl, and
the like. Suitable bases which can be used include DCC, TEA, and the like. The
reaction is
typically conducted in a polar organic solvent such as methanol or DMF. The
reaction is
typically conducted at low temperatures in the range of -10°C to
20°C.
Pharmaceutical Formulations
[0108] When employed as pharmaceuticals, the compounds of the subject
invention are
usually administered in the form of pharmaceutical compositions. These
compounds can be
administered by a variety of routes including oral, parenteral, transdermal,
topical, rectal, and
intranasal. These compounds are effective as both injectable and oral
compositions. Such
compositions are prepared in a manner well known in the pharmaceutical art and
comprise at
least one active compound.
32



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0109] This invention also includes pharmaceutical compositions that contain,
as the active
ingredient, one or more of the compounds of the subject invention above
associated with
pharmaceutically acceptable carriers. In making the compositions of this
invention, the active
ingredient is usually mixed with an excipient, diluted by an excipient or
enclosed within such a
carrier which can be in the form of a capsule, sachet, paper or other
container.. The excipient
employed is typically an excipient suitable for administration to human
subjects or other
mammals. When the excipient serves as a diluent, it can be a solid, semi-
solid, or liquid
material, which acts as a vehicle, carrier or medium for the active
ingredient. Thus, the
compositions can be in the form of tablets, pills, powders, lozenges, sachets,
cachets, elixirs,
suspensions, emulsions, solutions, syrups, aerosols, (as a solid or in aliquid
medium), ointments
containing, for example, up to 10% by weight of the active compound, soft and
hard gelatin
capsules, suppositories, sterile injectable solutions, and sterile packaged
powders.
[0110] In preparing a formulation, it may be necessary to mill the active
compound to
provide the appropriate particle size prior to combining with the other
ingredients. If the active
compound is substantially insoluble, it ordinarily is milled to a particle
size of less than 200
mesh. If the active compound is substantially water soluble, the particle size
is normally
adjusted by milling to provide a substantially uniform distribution in the
formulation, e.g., about
40 mesh.
[0111] Some examples of suitable excipients include lactose, dextrose,
sucrose, sorbitol,
mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth,
gelatin, calcium
silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile
water, syrup, and
methyl cellulose. The formulations can additionally include: lubricating
agents such as talc,
magnesium stearate, and mineral oil; wetting agents; emulsifying and
suspending agents;
preserving agents such as methyl- and propylhydroxy-benzoates; sweetening
agents; and
flavoring agents. The compositions of the invention can be formulated so as to
provide quick,
sustained or delayed release of the active ingredient after administration to
the patient by
employing procedures known in the art.
[0112] The quantity of active component, that is the compound according to the
subject
invention, in the pharmaceutical composition and unit dosage form thereof may
be varied or
adjusted widely depending upon the particular application, the potency of the
particular
compound and the desired concentration.
[0113] The compositions are preferably formulated in a unit dosage form, each
dosage
containing from about 5 to about 100 mg, more usually about 10 to about 30 mg,
of the active
33



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
ingredient. The term "unit dosage forms" refers to physically discrete units
suitable as unitary
dosages for human subjects and other mammals, each unit containing a
predetermined quantity
of active material calculated to produce the desired therapeutic effect, in
association with a
suitable pharmaceutical excipient. Preferably, the compound of the subject
invention above is
employed at no more than about 20 weight percent of the pharmaceutical
composition, more
preferably no more than about 15 weight percent, with the balance being
pharmaceutically inert
carrier(s).
[0114] The active compound is effective over a wide dosage range and is
generally
administered in a pharmaceutically or therapeutically effective amount. It
will be understood,
however, that the amount of the compound actually administered will be
determined by a
physician, in the light of the relevant circumstances, including he condition
to be treated, the
severity of the bacterial infection being treated, the chosen route of
administration, the actual
compound administered, the age, weight, and response of the individual
patient, the severity of
the patient's symptoms, and the like.
[0115] In therapeutic use for treating, or combating, bacterial infections in
warm-blooded
animals, the compounds or pharmaceutical compositions thereof will be
administered orally,
topically, transdermally, and/or parenterally at a dosage to obtain and
maintain a concentration,
that is, an amount, or blood-level of active component in the animal
undergoing treatment which
will be antibacterially effective. Generally, such antibacterially or
therapeutically effective
amount of dosage of active component (i.e., an effective dosage) will be in
the range of about
0.1 to about 100, more preferably about 1.0 to about 50 mg/kg of body
weight/day.
[0116] For preparing solid compositions such as tablets, the principal active
ingredient is
mixed with a pharmaceutical excipient to form a solid preformulation
composition containing a
homogeneous mixture of a compound of the present invention. When referring to
these
preformulation compositions as homogeneous, it is meant that the active
ingredient is dispersed
evenly throughout the composition so that the composition may be readily
subdivided into
equally effective unit dosage forms such as tablets, pills and capsules. This
solid preformulation
is then subdivided into unit dosage forms of the type described above
containing from, for
example, 0.1 to about 500 mg of the active ingredient of the present
invention.
[0117] The tablets or pills of the present invention may be coated or
otherwise compounded
to provide a dosage form affording the advantage of prolonged action. For
example, the tablet
or pill can comprise an inner dosage and an outer dosage component, the latter
being in the form
of an envelope over the former. The two components can be separated by an
enteric layer that
34



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
serves to resist disintegration in the stomach and permit the inner component
to pass intact into
the duodenum or to be delayed in release. A variety of materials can be used
for such enteric
layers or coatings, such materials including a number of polymeric acids and
mixtures of
polymeric acids with such materials as shellac, cetyl alcohol, and cellulose
acetate.
[0118] The liquid forms in which the novel compositions of the present
invention may be
incorporated for administration orally or by injection include aqueous
solutions, suitably
flavored syrups, aqueous or oil suspensions, and flavored emulsions with
edible oils such as
corn oil, cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as
elixirs and similar
pharmaceutical vehicles.
[0119] Compositions for inhalation or insufflation include solutions and
suspensions in
pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof,
and powders.
The liquid or solid compositions may contain suitable pharmaceutically
acceptable excipients as
described supra. Preferably the compositions are administered by the oral or
nasal respiratory
route for local or systemic effect. Compositions in preferably
pharmaceutically acceptable
solvents may be nebulized by use of inert gases. Nebulized solutions may be
inhaled directly
from the nebulizing device or the nebulizing device may be attached to a face
mask tent, or
intermittent positive pressure breathing machine. Solution, suspension, or
powder compositions
may be administered, preferably orally or nasally, from devices that deliver
the formulation in an
appropriate manner.
[0120] The following formulation examples illustrate representative
pharmaceutical
compositions of the present invention.
Formulation Example 1
[0121] Hard gelatin capsules containing the following ingredients are
prepared:
uanti


In,_reg diem (mg/capsule)


Active Ingredient 30.0


Starch 3 05.0


Magnesium stearate 5.0


[0122] The above ingredients are mixed and filled into hard gelatin capsules
in 340 mg
quantities.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Formulation Example 2
[0123] A tablet formula is prepared using the ingredients below:
uanti


In reg client ~ (capsule)


Active Ingredient


25.0


Cellulose, microcrystalline200.0


Colloidal silicon dioxide10.0


Stearic acid 5.0


[0124] The components are blended and compressed to form tablets, each
weighing 240 mg.
Formulation Example 3
[0125] A dry powder inhaler formulation is prepared containing the following
components:
Ingredient Wei hg t
Active Ingredient 5
Lactose 95
[0126] The active ingredient is mixed with the lactose and the mixture is
added to a dry
powder inhaling appliance.
Formulation Example 4
[0127] Tablets, each containing 30 mg of active ingredient, are prepared as
follows
uanti


In re,~ client (m~/capsule)


Active Ingredient 30.0 mg


Starch 45.0 mg


Microcrystalline cellulose 35.0 mg


Polyvinylpyrrolidone


(as 10% solution in sterile water)4.0 mg


Sodium carboxymethyl starch 4.5 mg


Magnesium stearate 0.5 mg


Talc 1.0 mg


Total 120 mg


[0128] The active ingredient, starch and cellulose are passed through a No. 20
mesh U.S.
sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with
the resultant
powders, which are then passed through a 16 mesh U.S. sieve. The granules so
produced are
dried at 50°C to 60°C and passed through a 16 mesh U.S. sieve.
The sodium carboxymethyl
starch, magnesium stearate, and talc, previously passed through a No. 30 mesh
U.S. sieve, are
36



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
then added to the granules which, after mixing, are compressed on a tablet
machine to yield
tablets each weighing 120 mg.
Formulation Example 5
[0129] Capsules, each containing 40 mg of medicament are made as follows:
uanti


In. re~~ diem (m~/capsulel


Active Ingredient 40.0 mg


Starch 109.0 mg


Magnesium stearate 1.0 mg


Total 150.0 mg


[0130] The active ingredient, starch and magnesium stearate are blended,
passed through a
No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg
quantities.
Formulation Example 6
[0131] Suppositories, each containing 25 mg of active ingredient are made as
follows:
Ingredient Amount
Active Ingredient 25
Saturated fatty acid glycerides to 2,000 mg
[0132] The active ingredient is passed through a No. 60 mesh U.S. sieve and
suspended in
the saturated fatty acid glycerides previously melted using the minimum heat
necessary. The
mixture is then poured into a suppository mold of nominal 2.0 g capacity and
allowed to cool.
Formulation Example 7
[0133] Suspensions, each containing 50 mg of medicament per 5.0 mL dose are
made as
follows:
In reg client Amount


Active Ingredient 50 mg


Xanthan gum 4.0 mg


Sodium carboxymethyl cellulose
(11%)


Microcrystalline cellulose (89%)50.0 mg


Sucrose 1.75 g


Sodium benzoate 10.0 mg


Flavor and Color q.v.


Purified water to 5.0 mL


[0134] The active ingredient, sucrose and xanthan gum are blended, passed
through a No. 10
mesh U.S. sieve, and then mixed with a previously made solution of the
microcrystalline
cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate,
flavor, and color
37



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
are diluted with some of the water and added with stirring. Sufficient water
is then added to
produce the required volume.
Formulation Example 8
uanti


Ingredient (m~gl''~capsule~


Active Ingredient 15.0 mg


Starch 407.0 mg


Magnesium stearate 3.0 mg


Total 425.0 mg


[0135] The active ingredient, starch, and magnesium stearate are blended,
passed through a
No. 20 mesh U.S. sieve, and filled into hard.gelatin capsules in 425.0 mg
quantities.
Formulation Example 9
[0136] A subcutaneous formulation may be prepared as follows:
Ingredient uanti
Active Ingredient 5.0 mg
Corn Oil 1.0 mL
Formulation Example 10
[0137] A topical formulation may be prepared as follows:
Ingredient uanti


Active Ingredient 1-10 g


Emulsifying Wax 30 g


Liquid Paraffin 20 g


White Soft Paraffin to 100 g


[0138] The white soft paraffin is heated until molten. The liquid paraffin and
emulsifying
wax are incorporated and stirred until dissolved. The active ingredient is
added and stirring is
continued until dispersed. The mixture is then cooled until solid.
Formulation Example 11
[0139] An intravenous formulation may be prepared as follows:
Ingredient uanti
Active Ingredient 250 mg
Isotonic saline 1000 mg
[0140] Another preferred formulation employed in the methods of the present
invention
employs transdermal delivery devices ("patches"). Such transdermal patches may
be used to
provide continuous or discontinuous infusion of the compounds of the present
invention in
38



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
controlled amounts. The construction and use of transdermal patches for the
delivery of
pharmaceutical agents is well known in the art. See, e.g., U.S. Patent
5,023,252, issued June 11,
1991, herein incorporated by reference. Such patches may be constructed for
continuous,
pulsatile, or on demand delivery of pharmaceutical agents.
[0141] Frequently, it will be desirable or necessary to introduce the
pharmaceutical
composition to the brain, either directly or indirectly. Direct techniques
usually involve
placement of a drug delivery catheter into the host's ventricular system to
bypass the blood-
brain barrier. One such implantable delivery system used for the transport of
biological factors
to specific anatomical regions of the body is described in U.S. Patent
5,011,472 which is herein
incorporated by reference.
[0142] Indirect techniques, which are generally preferred, usually involve
formulating the
compositions to provide for drug latentiation by the conversion of hydrophilic
drugs into lipid-
soluble drugs. Latentiation is generally achieved through blocking of the
hydroxy, carbonyl,
sulfate, and primary amine groups present on the drug to render the drug more
lipid soluble and
amenable to transportation across the blood-brain barrier. Alternatively, the
delivery of
hydrophilic drugs may be enhanced by infra-arterial infusion of hypertonic
solutions which can
transiently open the blood-brain barrier.
[0143] Other suitable formulations for use in the present invention can be
found in
Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia,
PA, 17th ed.
(1985).
[0144] As noted above, the compounds described herein are suitable for use in
a variety of
drug delivery systems described above. Additionally, in order to enhance the
in vivo serum half
life of the administered .compound, the compounds may be encapsulated,
introduced into the
lumen of liposomes, prepared as a colloid, or other conventional techniques
may be employed
which provide an extended serum half life of the compounds. A variety of
methods are
available for preparing liposomes, as described in, e.g., Szoka, et al., U.S.
Patent Nos.
4,235,871, 4,501,728 and 4,837,028 each of which is incorporated herein by
reference. .
[0145] As noted above, the compounds administered to a patient are in the form
of
pharmaceutical compositions described above. These compositions may be
sterilized by
conventional sterilization techniques, or may be sterile filtered. The
resulting aqueous solutions
may be packaged for use as is, or lyophilized, the lyophilized preparation
being combined with a
sterile aqueous carrier prior to administration. The pH of the compound
preparations typically
will be between 3 and 1 l, more preferably from 5 to 9 and most preferably
from 7 and 8. It will
39



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
be understood that use of certain of the foregoing excipients, carriers, or
stabilizers will result in
the formation of pharmaceutical salts.
[0146] In general, the compounds of the subject invention will be administered
in a
therapeutically effective amount by any of the accepted modes of
administration for agents that
serve similar utilities. Toxicity and therapeutic efficacy of such compounds
can be determined
by standard pharmaceutical procedures in cell cultures or experimental
animals, e.g., for
determining the LDSO (the dose lethal to 50% of the population) and the EDSO
(the dose
therapeutically effective in 50% of the population). The dose ratio between
toxic and
therapeutic effects is the therapeutic index and it can be expressed as the
ratio LDSO/ EDso.
Compounds that exhibit large therapeutic indices are preferred.
[0147] The data obtained from the cell .culture assays and animal studies can
be used in
formulating a range of dosage for use in humans. The dosage of such compounds
lies preferably
within a range of circulating concentrations that include the EDSO with little
or no toxicity. ~ The
dosage may vary within this range depending upon the dosage form employed and
the route of
administration utilized. For any compound used in the method of the invention,
the
therapeutically effective dose can be estimated initially from cell culture
assays. A dose may be
formulated in animal models to achieve a circulating plasma concentration
range which includes
the ICSO (the concentration of the test compound which achieves a half maximal
inhibition of
symptoms) as determined in cell culture. Such information can be used to more
accurately
determine useful doses in humans. Levels in plasma may be measured, for
example, by high
performance liquid chromatography.
LTtili
[0148] The compounds, prodrugs and pharmaceutically acceptable salts thereof,
as defined
herein, may have activity against a variety of bacteria, protozoa, fungi, and
parasites. By way of
example, the compounds, prodrugs and pharmaceutically acceptable salts thereof
may be active
against gram positive and gram negative bacteria. The compounds, prodrugs and
pharmaceutically acceptable salts thereof may be active against a variety of
fungi, including
fungi from the genus Mucor and Candida, e.g., Mucor racemosus or Cahdida
albicans. The
compounds, prodrugs and pharmaceutically acceptable salts thereof may be
active against a
variety of parasites, including malaria and cyptospo~idium paxasite.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0149] The compounds of the subject invention exhibit activity against a
variety of bacterial
infections, including, for example, gram positive infections, gram negative
infections,
mycobacteria infections, mycoplasma infections, and chlamydia infections.
[0150] Since the compounds of the subject invention exhibit potent activities
a variety of
bacteria, such as gram positive bacteria, the compounds of the present
invention are useful
antimicrobial agents and may be effective against a number of human and
veterinary pathogens,
including gram positive bacteria. The Gram positive organisms against which
the compounds of
the present invention are effective include Streptococcus pneumoniae,
Staphylococcus aureus,
Staphylococcus epidermidis, Enterococcus faecalis, Enterococcus faecium,
Haemophilus
influenzae, Mo~axella catarrhalis, Escherichia coli, Bacte~oides fragilis,
Bacte~oides
thetaiotaomicron, and Clostridium difficile, and the like.
[0151] The compounds of the subject invention may be combined with one or more
additional antibacterial agents. One or more of the additional antibacterial
agents may be active
against gram negative bacteria. Additionally, one or more of the additional
antibacterial agents
may be active against gram positive bacteria. The combination of the compounds
of the subject
invention and the one or may additional antibacterial agents may be used to
treat a gram
negative infection. Additionally, the combination of the compounds of the
subject invention and
the one or more additional antibacterial agents may be used to treat a gram
positive infection.
The combination of compounds of the subject invention and the one or more
additional
antibacterial agents may also be used to treat a mycobacteria infection,
mycoplasma infection, or
chlamydia infection.
[0152] The in vitro activity of compounds of the subject invention may be
assessed by
standard testing procedures such as the determination of minimum inhibitory
concentration
(MIC) by agar dilution as described in "Approved Standard. Methods for
Dilution
Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically," 3rd
ed., published 1993
by the National Committee for Clinical Laboratory standards, Villanova,
Pennsylvania, USA.
[0153] The amount administered to the mammalian patient will vary depending
upon what is
being administered, the purpose of the administration, such as prophylaxis or
therapy, the state
of the patient, the manner of administration, and the like. In therapeutic
applications,
compositions are administered to a patient already suffering from a disease in
an amount
sufficient to cure or at least partially arrest the symptoms of the disease
and its complications.
An amount adequate to accomplish this is defined as "therapeutically effective
dose." Amounts
effective for this use will depend on the disease condition being treated as
well as by the
41



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
judgment of the attending clinician depending upon factors such as the
severity of the
inflammation, the age, weight and general condition of the patient, and the
like.
[0154] The compositions administered to a patient are in the form of
pharmaceutical
compositions described above. These compositions may be sterilized by
conventional
sterilization techniques, or may be sterile filtered. The resulting aqueous
solutions may be
packaged for use as is, or lyophilized, the lyophilized preparation being
combined with a sterile
aqueous carrier prior to administration. The pH of the compound preparations
typically will be
between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8.
It will be
understood that use of certain of the foregoing excipients, carriers, or
stabilizers will result in the
formation of pharmaceutical salts.
[0155] The therapeutic dosage of the compounds of the present invention will
vary
according to, for example, the particular use for which the treatment is made,
the manner of
administration of the compound, the health and condition of the patient, and
the judgment of the
prescribing physician. For example, for intravenous administration, the dose
will typically be in
the range of about 20 mg to about 500 mg per kilogram body weight, preferably
about 100 mg
to about 300 mg per kilogram body weight. Suitable dosage ranges for
intranasal
administration are generally about 0.1 mg to 1 mg per kilogram body weight.
Effective doses
can be extrapolated from dose-response curves derived from in vitro or animal
model test
systems.
[0156] The following synthetic and biological examples are offered to
illustrate this
invention and are not to be construed in any way as limiting the scope of this
invention.
EXAMPLES
[0157] In the discussion above and in the examples below, the following
abbreviations have
the following meanings. If an abbreviation is not defined, it has its
generally accepted meaning.
42



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
7-methyl/MTL - 1-methylthio-7-deoxy-7-


methyllincosamine


apt - apparent triplet


a~n - atmospheres


Bn - benzyl


Boc - test-butoxycarbonyl protecting
group


br s - broad singlet


BSTFA - N, O-


bis(trimethylsilyl)trifluoroacetamide


Cbz - carbonyloxybenzyloxy protecting
group


CDC13 - deuterated chloroform


CD30D - deuterated methanol


cfu - colony forming units


D - doublet


DAST - dimethylaminosulfurtrifluoride


dd - doublet of doublets


dddd - doublet of doublets of doublet
of


doublets


dt - doublet of triplets


DCE - dicholoroethane


DCM - dichloromethane


DIEA - diisopropyethylamine


DMAP - dimethylaminopyridine


DMF - dimethylformamide


DMSO - dimethyl sulfoxide


DPPA - diphenylphosphoryl azide


EDSO - dose therapeutically effective
in 50% of


the population


Equiv - equivalents


ESMS - electrospray mass spectrometry


Et - ethyl


EtOAc - ethyl acetate


Et20 - diethyl ether


g - grams


h - hours


HATU - O-(7-azabenzotriazol-1-yl)-N,N,N',N'-


tetramethyluronium


hexafluorophosphate


HOBT - 1-hydroxybenzotriazole hydrate


1H NMR - Hydrogen Nuclear Magnetic Resonance


spectroscopy


HPLC - high pressure liquid chromatography


Hz - hertz


ICSO - concentration of the test compound


which achieves a half maximal


inhibition of symptoms


,I - coupling constant in hertz


L - liters


LDSO - dose lethal to 50% of the population


43



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
LiHMDS - lithium hexamethyldisilazide


LiN(iPr)2 - lithium diisopropylnitride


m - multiplet


M - molar


MCPBA - 2-(4-chloro-o-tolyloxy) acetic
acid


Me - methyl


MeCN - acetonitrile


MeOH - methanol


mg - milligrams


MHz - megahertz


Min - minutes


mL - milliliters


millimeter


mmol - millimol


MS(ESPOS) - mass spectrometry by positive
mode


electrospray ionization


MS(ESNEG) - Mass Spectrometry by negative
mode


electrospray ionization


MTL - 1-methylthiolincosamine (methyl
6-


amino-6,8-dideoxy-1-thio-erythro-a-D-


galacto-octopyranoside)


N - normal


NMR - nuclear magnetic resonance


OBz - benzyloxy protecting group


OtBu - tert-butoxy


Pd/C - palladium/carbon


pg - picograms


ph - phenyl


Pro ' - L-proline


psi pounds per square inch


q - quartet


q,v, - quantitative


Rf - Retention factor


- room temperature


s - singlet


sat. - saturated


t - triplet


TEA - triethylamine


TFA - trifluoroacetic acid


THF - tetrahydrofuran


TLC - thin layer chromatography


~,g - micrograms


~,L - microliters


~,m - micromolar


v/v - volume by volume


w/w - weight by weight


[0158] Additionally, the term "Aldrich" indicates that the compound or reagent
used in the
following procedures is commercially available from Aldrich Chemical Company,
Inc., 1001
44



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
West Saint Paul Avenue, Milwaukee, WI 53233 USA; the term "Fluka" indicates
that the
compound or reagent is commercially available from Fluka Chemical Corp., 980
South 2nd
Street, Ronkonkoma NY 11779 USA; the term "Lancaster" indicates that the
compound or
reagent is commercially available from Lancaster Synthesis, Inc., P.O. Box 100
Windham, NH
03087 USA; the term "Sigma" indicates that the compound or reagent is
commercially available
from Sigma, P.O. Box 14508, St. Louis MO 63178 USA; the term "Chemservice"
indicates
that the compound or reagent is commercially available from Chemservice Inc.,
Westchester,
PA, USA; the term "Bachem" indicates that the compound or reagent is
commercially available
from Bachem Bioscience Inc., 3700 Horizon Drive, Renaissance at Gulph Mills,
King of
Prussia, PA 19406 USA; the term "Maybridge" indicates that the compound or
reagent is
commercially available from Maybridge Chemical Co.
[0159] Trevillett, Tintagel, Cornwall PL34 OHW United Kingdom; the term "RSP"
indicates that the compound or reagent is commercially available from RSP
Amino Acid
Analogs, Inc., 106 South St., Hopkinton, MA 01748, USA, and the term "TCI"
indicates that the
compound or reagent is commercially available from TCI America, 9211 North
Harborgate St.,
Portland, Oregon, 97203, OR, USA; the term "Toronto" indicates that the
compound or reagent
is commercially available from Toronto Reasearch Chemicals, Inc., 2 Brisbane
Rd., New York,
ON, Canada M3J2J8; the term "Alfa" indicates that the compound or reagent is
commercially
available from Johnson Matthey Catalog Company, Inc. 30 Bond Street, Ward
Hill, MA
018350747; and the term "Nova Biochem" indicates that the compound or reagent
is
commercially available from NovaBiochem USA, 10933 North Torrey Pines Road,
P.O. Box
12087, La Jolla CA 92039-2087.
[0160] In the examples below, all temperatures are in degrees Celsius (unless
otherwise
indicated) and the following general procedures are used to prepared the
compounds as
indicated. It will be appreciated by one of skill in the art that the
following general procedures
are meant to be illustrative only and that the methods may be broadened to
synthesize other
compounds of the subject invention.
General Procedures
Method A
[0161] Methyl 6-amino-6,8-dideoxy-1-thio-erythro-a-D-galacto-octopyranoside la
(Rl = Me) (MTL) was prepared as described by Hoeksema, H. et al., J. Am. Chem.
Soc.,1967,



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
89, 2448-2452. N-(Benzyloxycarbonyloxy)succinimide (5.8 g 23.1 mmol) and la
(5.0 g, 19.7
mmol) were suspended in pyridine (40mL) and stirred under N2 atmosphere 36 h.
The reaction
mixture was cooled to 0°C and then bis-N, O-trifluoroacetamide (15.7
mL, 59.0 mmol) was
added by syringe over 2 min. The reaction mixture was allowed to warm to rt
and stirred for 42
h. Toluene (100 mL) was added and the reaction mixture was evaporated to
dryness. The
residue was taken up in ethyl acetate (400 mL). The organic solution was
washed quickly with
10% citric acid (200 mL), Ha0 (3 x 100 mL), saturated NaHCO3 (100 mL), and
brine (2 x 100
mL), and dried over NaaS04 and evaporated to dryness. Chromatography of the
crude product
on silica 10% EtOAc/Hexanes containing 0.2% TEA after co-evaporation from
toluene (100
mL) and cyclohexane (2 x 100 mL) provided the protected product lb (P=Cbz,
Rl=Me) (7.2 g,
54%) as a colorless oil.
[0162] 1H NMR (300 MHz, CD3SOCD3) 8 7.34-7.31 (m, 5), 7.05 (d, J 8.2, 1), 5.19
(d, J--
5.8, 1), 5.01 (d, J--1.6, 2), 3.99 (apt dt, J-- 5.5, 9.3, 9.3, 2), 3.93-3.86
(m, 3), 3.49 (dd, J-- 2.5,
9.6, 1), 2.01 (s, 3), 1.03 (d, J 6.3, 3), 0.10 (s, 9), 0.09 (s, 9), 0.04 (m,
18).
[0163] To dimethylsulfoxide (413 p.L, 5.82 mmol) in DCM (1.5 mL) cooled to -
72°C was
added oxalyl chloride 2 M in DCM (1.49 mL, 2.98 mmol) over 1 min. After 25 min
the
protected product lb (1.92 g, 2.84 mmol) in DCM (4.0 mL) was added by cannula.
The
resulting reaction mixture was stirred for 25 min and then allowed to warm to -
50°C (dry ice
acetonitrile) and maintained at this temperature for 2 h. To the reaction
mixture was added TEA
(1.29 mL, 3.30 mmol). After 25 min the reaction mixture was diluted with EtOAc
(300 mL).
The resulting organic solution was washed quickly with 5% citric acid (300
mL), HZO (2 x
300 mL), saturated NaHC03 (100 mL), brine (100 mL) dried over Na2SO4 and
evaporated to
dryness with the aid of toluene (100 mL) to provide the product lc. The
product lc (P=Cbz,
Rl=Me) was obtained as a colorless crystalline solid (1.60 g, 94%) after co-
evaporation with
n-pentane and removal of residual solvent under high vacuum.
[0164] 1H NMR (300 MHz CDC13) 8 7.37-7.33 (m, 5), 5.60 (m, 1), 5.21 (d, J=
5.2, 1), 5.17
(d, J 12.4, 1 ), 5.08 (d, J-- 12.4, 1 ), 4.74 (m, 1 ), 4.16-4.12 (m, 2), 3.87
(d, J = 2.2, 1 ), 3 .69 (dd,
J 2.5, 9.3, 1), 2.01 (br s, 3), 1.90 (s, 3), 0.19 (s, 9), 0.16 (s, 9), 0.15
(s, 9).
Method B
[0165] The Boc-protected product lc (P=Boc, Rl =Me) may be prepared in general
as
outlined above. la (Rl = Me) (MTL) (Dried at 50°C high vacuum) (21.8 g,
86 mmol) was
suspended in methanol (200 mL) and TEA (26 mL) and was cooled to 0°C on
ice. To this
46



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
mixture di-t-butyldicarbonate (57.0 g, 0.26 mol) was added. The reaction
mixture was then
stirred overnight at rt. To the reaction mixture was added toluene (100 mL).
The solvents were
removed to a total volume of 100 mL, leaving a thick suspension to which
cyclohexane (300
mL) was added. The resulting solid precipitate was triturated, then filtered
and washed with
cyclohexane, ether, and pentane and dried to constant weight. The crude Boc-
protected product
was used without further purification (87%).
[0166] TLC R~= 0.75 (10% MeOH/DCM); MS(ESPOS): 354 (M+H); 1H NMR (300 MHz,
CD3OD) 8 0.14 (d, J= 6.3, 3),1.43 (s, 9), 2.07 (s, 3), 3.55 (dd, J= 3.3,
10.43, 1), 3.84 -4.08 (m,
3), 4.10-4.15 (m, 2), 5.25 (d, J= 5.5, 1).
[0167] To N Boc-1-methylthiolincosamide (240 mg, 0.68 mmol) in DMF (5 mL,
BSTFA
(0.52 mL, 2.0 mmol) and triethylamine (0.14 mL, 1.42 mmol) were added at
0°C and then
stirred at rt overnight. DMF was removed and the crude product was quickly
passed through a
silica gel column (pretreated with 2% TEA in ethyl acetate) eluting with 10%
ethyl acetate in
hexanes lb (P=Boc, Rl=Me) (350 mg, 95%). To oxalyl chloride (0.16 mL, 0.78
mmol) in
dichloromethane (5 mL) at -60°C, dimethylsulfoxide (0.22 mL, 0.78 mmol)
was added slowly
and then stirred for 15 min. After which, lb (370 mg, 0.65 mmol) in DCM (5 mL)
was added
slowly. The reaction mixture was stirred for 45 min, during which the reaction
temperature was
raised to -40°C. Triethylamine (0.70 mL, 3.25 mmol) was then added and
the stirring continued
for an additional 15 min at -40°C. It was then extracted with DCM (100
mL) and washed with
10% citric acid (50 mL). The residue obtained on removal of solvent was then
purified on silica
gel column using 10% ethyl acetate in hexanes as eluent lc (P=Boc, Rl=Me) as a
colorless oil
(289 mg, 78%).
[0168] TLC: Rf= 0.60 (10% EtOAc/Hexanes); MS(ESPOS): 590 (M+23);1H NMR (300
MHz, CDC13) 8 0.11 (s, 18), 0.17 (s, 18), 1.40 (s, 9), 1.84 (s, 3), 2.26 (s,
3), 3.63 (dd, J= 2.7,
9.34, 1), 3.82 (d, J= 1.9, 1), 4.01-4.12 (m, 2), 5.15 (d, J= 5.5, 1).
Method C
[0169] Triphenylphosphonium bromide (3.29 g, 9.2 mmol) and potassium tent-
butoxide (715
mg, 6.4 mmol) under Na atmosphere were suspended in toluene (31 mL) with
vigorous stirring.
After 4 h protected product lc (P=Cbz, Rl=Me) (1.4 g, 2.36 mmol) in toluene
(20 mL) was
added by cannula. The resulting reaction mixture was stirred 2 h and then
diluted with EtOAc
(250 mL). The resulting organic solution was washed quickly with H2O (2 x 100
mL), brine (1
x 100 mL) dried over Na2S04 and evaporated to dryness. Chromatography of the
crude product
47



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
on silica 6% EtOAc/Hexanes containing 0.2% TEA gave the allcene product 2a
(P=Cbz, Ri=Me,
Ra=CH2) as a colorless oil that crystallized after co-evaporation from toluene
and cyclohexane
(0.65 g, 46%).
[0170] 1H NMR (300 MHz CDC13) ~ 7.35-7.27 (m, 5), 6.36 (d, J= 7.1, 1), 5.24
(d, J= 5.5,
1), 5.08 (m, 4), 4.34 (m, 1), 4.16 (m, 2), 3.88 (d, J= 2.2, 1), 3.61 (dd, J=
2.2, 9.3, 1), 2.20 (s, 3),
1.79 (s, 3), 0.17-0.13 (m, 27).
[0171] The product 2a (P=Cbz, Rl=Me, R2=CH2) (490 mg, 0.82 mmol) in ethanol
(50 mL)
was added to 10% palladium on carbon (Degussa wet form 50% w/w water) (700 mg)
in a par
bottle. The bottle was purged, and charged with HZ to 65 psi and shaken 24 h.
The reaction
mixture was filtered through celite, rinsed with methanol. The organic
solution was transferred
to a resin funnel containing dry, washed Dowex SOw-400x H+ form (0.8 g) and
shaken for 10
min. After washing the resin with methanol three times and water two times,
the saturated
product 2b (Rl =Me, R2=Me) was eluted from the resin by washing with 5% TEA in
MeOH (35
mL, x 10 min x 5). The combined filtrate was evaporated to dryness, co-
evaporated from EtOH
twice and lyophilized from 1:1 MeCNlHzO to give the product as a colorless
powder (198.4 mg
96%).
[0172] 'H NMR (300 MHz, Da0) 8 5.17 (d, J= 5.8, 1), 3.97-3.84 (m, 3), 3.52
(dd, J= 3.0,
10.0, 1), 2.82 (dd, J= 4.4, 8.5, 1), 1.94 (s, 3),1.89-1.81 (m, 1), 0.82 (d, J=
6.9, 3), 0.72 (d, J=
6.9, 3). MS(ESPOS): 252.2 [M+H], (ESNEG): 250.4 [M-H].
Method D
[0173] In the alternative when a Boc-protecting group is used,
methyltriphenylphosphonium
bromide (12 g, 33.6 mmol) and potassium t-butoxide (3g, 26.7 mmol) were taken
in THF (70
mL) at 0°C, and stirred at rt for 4 h. Then Boc-protected product lc
(P=Boc, Rl=Me) (4.7 g, 8.2
mmol) in THF (30 mL) was added and stirred at rt for 2 h. After which it was
extracted with
EtOAc (300 mL), washed with brine (100 mL) and dried over sodium sulfate. The
crude alkene
product 2a (P=Boc, Rl=Me, R2=CHz) was purified on silica gel column
chromatography using
10% EtOAc in Hexane as eluent (4.1 g, 87.6%).
[0174] TLC: Rf= 0.5 (10% of EtOAc in Hexane): 1H NMR (300 MHz, GD30D) b 7.24
(m,
2), 5.22 (d, J= 5.7, 1), 4.21 (m, 1), 4.09 (m, 2), 3.87 (d, J= 2.4, 1), 3.60
(dd, J= 2.7, 9.3, 1),
1.99 (s, 3),1.76 (s, 3); 1.43 (s, 9); MS(ESPOS): 444 (M-2TMS+Na).
[0175] To the product 2a (P=Boc, Rl=Me, R2=CHa) in methanol (30 mL), Dowex H+
resin
(1 g) was added and stirred at rt for 1 h. The resin was filtered and the
product obtained on
48



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
removal of solvent (2.4 g, 6.8 mmol,) was taken in MeOH (30 mL). Pd/C (2.5 g)
was added and
hydrogenated at 55 psi overnight. The crude product obtained on filtering and
removal of
solvent was purified on silica gel column chromatography using 10% MeOH in DCM
to provide
Boc-protected 7-Methyl MTL as a white solid (2.06 g, 86%). TLC Rf= 0.5 (10% of
MeOH in
DCM).
[0176] 1H NMR (300 MHz, CD30D) 8 5.23 (d, J= 5.4, 1), 4.11 (m, 1), 3.97 (d,
J=10.2, 1),
3.84 (m, 1), 3.52 (m, 1), 2.08 (s, 3), 1.44 (s, 9), 1.14 (m, 1), 0.93 (d, J=
6.9, 3), 0.85 (d, J= 6.9,
3); MS(ESPOS): 351(M+H).
[0177] To the Boc-protected 7-Methyl MTL (150 mg, 0.43 mmol) in dichloroethane
(6 mL),
dimethylsulfide (0.16 mL, 2.5 mmol) was added, followed by TFA (2 mL), water
(0.16 mL) and
stirred at rt for 1 h. The solvent was removed to obtain the crude product 2b
(Rl=Me, R2=Me).
After purification on silica gel column chromatography using 30% MeOH in DCM
as eluent, the
product 2b (Rl=Me, R2=Me) was obtained identical in all respects to the
material obtained from
method C.
Method E
[0178] Sodium hydride (80 mg, 3.3 mmol) under N~, atmosphere was suspended in
THF (4
mL) with vigorous stirring. The suspension was cooled to -30°C and
diethyl(cyanomethyl)phosphonate (805 ~.L, 5.0 mmol) was added. After 30 min
protected
product lc (P=Cbz, Rl=Me) (1.0 g, 1.7 mmol) in THF (3 mL) was added by
cannula. The
resulting reaction mixture was stirred 4 h and then diluted with EtOAc (250
mL). The resulting
organic solution was washed quickly saturated aqueous NaHC03 (1 x 100 mL),
brine (1 x 50
mL) dried over Na2S04 and evaporated to dryness. Chromatography of the crude
product on
silica 6% EtOAc/Hexanes to 10% EtOAc/Hexanes containing 0.2% TEA gave the
protected
alkene product 2a (P=Cbz, Rl=Me, R2=CHCN) as a colorless oil (0.38 g, 37%).
MS(ESPOS):
625.5.2 [M+H], ES(NEC'r): 659.5 [M+Cl].
[0179] The product 2a (P=Cbz, Rl=Me, R2=CHCN) (180 mg, U.29 mmol) in ethanol
(15
mL) was added to 10% palladium on carbon (Degussa wet form 50% w/w water) (300
mg) in a
Parr bottle and concentrated HCl (29 ~.L) was added. The bottle was purged,
and charged with
H2 to 65 psi and shaken for 24 h. The reaction mixture was filtered through
celite, rinsed with
methanol. The organic solution was transferred to a resin funnel containing
dry, washed Dowex
SOw-400x ITS form (1 g) and shaken 10 min. After washing the resin with
methanol twice and
water, the saturated product 2b (Rl=Me, RZ=CHaCN) was eluted from the resin by
washing with
49



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
5% TEA in MeOH (20 mL x 20 min x 3) and MeCN (20 mL x 20 min). The combined
organic
filtrate was evaporated to dryness lyophilized from 1:1 MeCN/H20 to give the
product 2b
(Rl=Me, R2=CHaCN) as a colorless solid (70 mg, 91%). ES(NEG): 275.3 [M-H].
Method F
(0180] To the protected product lc (P=Cbz, Rl=Me) (0.75g, 1.3 mmol) in THF
(7.3 mL)
was added MeMgCI (3M) in THF (7.0 mL 2.1 mmol) at 0°C. Over 30 min the
reaction mixture
was warmed to 4°C and after 4 h the reaction mixture was quenched with
1:3 saturated aqueous
NH4C1/H20 (1 OmL). The quenched mixture was diluted to 100 mL with water 'and
extracted
with DCM (4 x 50 mL). The combined organic phase was dried and evaporated. The
residue
was dissolved in 1:2:4 H20/HOAc/THF (100 mL) and stirred for 20 h, and then
evaporated with
the aid of toluene (2 x 100 mL). Chromatography 10:1 to 10:2 DCM / MeOH gave
product 3a
(P=Cbz, Rl=Me, R2=lVle) (153 mg, 31%).
[0181] (ESNEG): 399.5 [M-H].
[0182] 3a (P=Cbz, Rl=Me, R~=Me) (79 mg, 0.2 mmol) in ethanol (10 mL) was added
to
10% palladium on carbon (Degussa wet form 50% w/w water) (400 mg) in a Parr
bottle. The
bottle was purged, and charged with H2 to 65 psi and shaken 6 h. The reaction
mixture was
filtered through celite, rinsed with methanol. The combined filtrate was
evaporated to dryness
and lyophilized from 1:1 MeCN/Ha0 to give the product 3b (Rl=Me, R2=Me) as a
colorless
powder (42 mg, 80%).
[0183] 1H NMR (300 MHz, Da0) 8 5.33 (d, J= 5.8,-1), 4.83-4.06 (m, 3), 3.65-
3.60 (m, 1),
3.06-3.03 (m, 1), 2.18 (s, 3), 1.30 (s, 3), 1.23 (s, 3). MS(ESPOS): 268.4
[M+H], MS(ESNEG):
266.2 [M-H].
Method G
[0184] To the Boc-protected product lc (P=Boc, Rl=Me) (100 mg, 0.18 mmol) in
methanol
(3 mL), O-trimethylsilylhydroxylamine (0.10 mL, 0.88 mmol) was added and
stirred at rt
overnight. The solvent was removed to obtain the crude Boc-protected product
4a (P=Boc,
Rl=Me, R~=H). To the crude product 4a (95 mg, 0.15 mmol), 30% trifluoroacetic
acid in
dichloroethane (10 mL) and dimethyl sulfide (0.5 mL) were added and stirred
for 1 h. The
solvent was removed and the product 4b (Rl=Me, R'=H) was taken as such for the
next step.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0185] TLC: Rf= 0.35 (10% MeOH/DCM); MS(ESPOS): 267 (M+H); 1H NMR (300 MHz,
CD30D) 8 1.96 (s, 3), 2.09 (s, 3), 3.58 (dd, J= 3.3, 10.2, 1), 3.90 (s, 1),
4.11 (dd, J= 5.7, 10.20,
1), 4.19 (d, J= 5.4, 1), 4.50 (d, J= 5.1, 1), 5.36 (d, J= 5.7, 1).
Method H
[0186] To the Boc-protected product lc (P=Boc, Rl=Me) (100 mg, 0.176 mmol) in
methanol
(4 mL) and water (1 mL), O-alkylhydroxylamine hydrochloride (for example, O-
methylhydroxylamine hydrochloride) (60 mg, 0.70 mmol) and sodium acetate (57
mg, 0.70
mmol) were added and heated at 80°C for 3 h and then stirred at rt
overnight. The solvent was
removed under high vacuum to obtain the crude Boc-protected product 4a (P=Boc,
Rl=H,
R7=Me). The crude product 4a was taken in 30% trifluoroacetic acid in
dichloroethane (10 mL),
dimethylsulfide (0.5 mL) and stirred for 1 h at rt. The solvent was removed
and the residue was
kept under high vacuum for 1 h and the product 4b (Rl=Me, R7=Me) was taken as
such for the
next step.
[0187] TLC: Rte- 0.63 (10% MeOH/DCM); MS(ESPOS): 281 (M+H). 1H NMR (300 MHz,
CD30D) 8 1.95 (s, 3), 2.08 (s, 3), 3.60 (dd, J= 3.3, 10.20, 1), 3.92 (s, 3),
4.13 (dd, J= 4.8,
10.20, 1), 4.49 (d, J= 1.2, 1), 5.38 (d, J= 5.4, 1).
Method I
[0188] To the Boc-protected product lc (P=Boc, Rl=Me) (500 mg, 0.88 mmol) in
THF (10
mL), tetrabutylammonium fluoride (2.5 mmol, 1 M in THF) was added and the
ieaction mixture
was stirred at rt for 1 h. The solvent was removed and the residue was
purified on silica gel
column using 5% methanol in dichloromethane as eluent. The product (111 mg,
0.31 mmol)
obtained from the column was then taken in a mixture of dichloromethane (3 mL)
and pyridine
(3 mL) to which acetic anhydride (0.5 mL, 10.6 mmol) and dimethylaminopyridine
(80 mg, 1.7
mmol) were added and stirred at rt overnight. The solvent was removed and the
crude product
was purified on silica gel column using 30% ethyl acetate in hexanes as eluent
to provide 5a
(P=Boc, Rl=Me) (58 mg, 38%).
(0189] TLC: Rf= 0.73 (50% EtOAc/Hexanes); MS(ESPOS): 500 (M'Na). 1H NMR (300
MHz, CDCl3) 8 1.38 (s, 9), 1.91 (s, 3), 1.98 (s, 3), 2.07 (s, 3), 2.18 (s, 3),
4.33 (m, 1), 4.72 (m,
1 ), 4.94 (m, 1 ), 5.21 (m, 2), 5 .45 (s, 1 ), 5.57 (m, 1 ).
51



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0190] To product Sa (P=Boc, Rl=Me) (158 mg, 0.331 mmol) in DCM (5 mL),
dimethylaminosulfurtrifluoride (732 p,L, 3.31 mmol) was added and stirred
overnight. More
DCM was added and the organic portion was washed with sodium bicarbonate. The
residue
obtained on removal of solvent was purified on silica gel column
chromatography using 20%
ethyl acetate in hexanes as eluent (100 mg, 60%) to provide the protected
product (P=Boc,
Rl=Me). The Boc-protected product was taken up in 30% trifluoroacetic acid in
dichloroethane
and dimethylsulfide and stirred for 1 h at rt. The solvent was removed to
provide the product 5b
(Rl=Me).
[0191] TLC: Rf= 0.63 (40% MeOH/Hexanes); MS(ESPOS): 522 (M+23). 1H NMR (300
MHz, CDC13) ~ 1.40 (s, 9),1.69 (t, J= 18.9, 3), 1.98 (s, 3), 2.08 (s, 6), 2.13
(s, 3), 4.22-4.30 (m,
11), 4.53 (dd, J= 10.9, 25.3, 1), 5.16-5.28 (m, 2), 5.52 (s, 1), 5.63 (d, J=
5.2, 1).
Method J
[0192] Enolization (LiHMDS) and alkylation of 7a with 4-bromo-2-methyl-2-
butenyl
afforded a mixture of diastereomers of the lactam 7b (R9'=2-methyl-2-butenyl)
(61 %) according
to the literature procedure by Zhang, R.; et al., Journal of the Americah
Chemical Society. 1998,
120, 3894-3902. Compound 7a is commercially available from vendors such as
Bachem.
Alternatively, 7a can be prepared by methods well known in the art for an
example see Baldwin,
et al.; Tet~ahedron,1989, 45, 7449-7468.
[0193] The lactam 7b was reduced to the pyrrolidine 7c (R9'=2-methyl-2-
butenyl) (70%) by
the two-step sequence involving superhydride reduction of the lactam to the
hemiaminal and the
subsequent reduction of the hemiaminal with Et3SiH/BF3~OEt2. The pyrrolidine
7c (R9'=2-
methyl-2-butenyl) (778 mg, 2.08 mmol), 10% palladium on carbon (230 mg), in
anhydrous
methanol (25 mL) was subjected to Parr hydrogenolysis at 50 psi for 5 h. The
reaction mixture
was filtered through a celite pad and washed several times with methanol. The
combined
washings and filtrate were evaporated to dryness, affording, without further
purification, a
colorless oil 7d (R9=2-methylbutyl).
[0194] TLC: Rf= 0.3 [Solvent system: DCM:hexanes:MeOH(6:5:1)]. MS(NEGATIVE):
284.5 [M-H].
52



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Method K
(0195] Enolization (LiHMDS, 33 mmol, 33 mL, 1.1 equiv) and alkylation of 7a
(9.47 g,
29.7 mmol, 1 equiv) with cis-1-bromo-2-pentene (4.21 mL, 35.6 mmol, 1.2
equiv), in anhydrous
THF at -78°C under nitrogen, afforded a mixture of diastereomers of the
lactam 7b (R9'=2-
pentenyl) (43.2%) after silica gel purification. The lactam 7b (3.96g,
10.22mmo1) was reduced
to the pyrrolidine 7c (R9'=2-pentenyl) by the two-step sequence involving
superhydride
reduction of the lactam to the hemiaminal, at -78°C in anhydrous THF,
and the subsequent
reduction of the herniaminal with Et3SiHBF30Et~ in anhydrous DCM at -
78°C affording 7c
(R9'=2-pentenyl) (71 %) after silica gel purification. The pyrrolidine 7c
(2.71 g, 7.26 mmol),
10% palladium on carbon (560mg), in anhydrous methanol (30 mL) was subjected
to Parr
hydrogenolysis at 50 psi for 5 h. The reaction mixture was filtered through a
celite pad and
washed several times with methanol. The combined washings and filtrate were
evaporated to
dryness, affording, without further purification, a colorless oil 7d
(R9=pentyl) (1.68g, 80%).
[0196] TLC: Rf= 0.3 [Solvent system:DCM:hexanes:MeOH(6:5:1)]. MS(ESNEG): 284.5
[M-H].
Method L
[0197] Ozonolysis of 7d (R9=2-methylbutyl) in anhydrous dichloromethane
followed by
treatment with DMS at -78°C afforded aldehyde 8a (77%). 4-Fluorobenzyl
phosphonium
chloride (0.87 g, 2.13 mmol) and potassium t-butoxide (0.17 g, 1.48 mmol) were
suspended in
toluene under nitrogen with vigorous stirring. After 4 h, a solution of
aldehyde 8a (204 mg, 0.59
mmol) in toluene (4.6 mL) was added drop-wise. The reaction mixture was
stirred at rt for 2 h
and diluted with ethyl acetate (50 mL). The organic layer was washed with
water (2 x 20 mL),
brine, dried and concentrated. The residue was purified by chromatography to
give a clear syrup
8b (R9'=3-(4-fluorophenyl)prop-2-enyl) (171 mg).
[0198] To a solution of 8b (R9~=3-(4-fluorophenyl)prop-2-enyl) (171 mg, 0.39
mmol) in
MeOH (25 mL) in a Parr bottle was added 10% palladium on carbon (Degussa wet
form 50%
w/w water) (200 mg). The bottle was purged and charged with H2 to 40 psi, and
shaken for 4 h.
The reaction mixture was filtered through celite and rinsed with MeOH. The
filtrate was
concentrated to give a yellow oil 8c (R9=3-(4-fluorophenyl)propyl) (120 mg).
[0199] MS(ESPOS): 374.5 [M +Na]+, MS(ESNEG): 350.3 [M - H] .
53



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Method M
[0200] 4-Chlorobenzyl phosphonium chloride (0.95g, 2.24 mmol, 3.9 equiv) and
potassium
t-butoxide (0.17 g, 1.55 mmol, 2.7 equiv) were suspended in toluene (7.5 mL)
under nitrogen
with vigorous stirring. After 4 h, a solution of aldehyde 8a (200 mg, 0.58
mmol, 1 equiv) in
toluene (4.9 mL) was added dropwise. The reaction mixture was stirred at rt
for 2 h and diluted
with ethyl acetate (50 mL). The organic layer was washed with water (2 x 20
mL), brine, dried
and concentrated. The residue was purified by chromatography to give a clear
syrup 8b (R9'=3-
(4-chlorophenyl)prop-2-enyl) (216 mg, 82%).
[0201] MS(ESPOS): 478.5 [M +Na]+, MS(ESNEG): 454.4 [M - H] .
[0202] To a solution of 8b (R9'=3-(4-chlorophenyl)prop-2-enyl) (147 mg, 0.32
mmol) in
cyclohexane (50 mL) was added 10% palladium on carbon (Degussa wet form 50%
w/w water)
(80 mg). The reaction mixture was stirred at rt under 1 atm H2 overnight. The
reaction mixture
was filtered through celite and rinsed with MeOH. The filtrate was
concentrated to give the
alkane product 8c (R9=3-(4-chlorophenyl)propyl) as a clear oil (131 mg, 89%).
To a solution of
the alkane (131 mg, 0.29 mmol, 1 equiv) in THF (3 mL) and water (1 mL) was
added lithium
hydroxide monohydrate (60 mg, 1.43 mmol, 5 equiv). The reaction mixture was
stirred at rt
overnight. The THF was removed under vacuum. The residue was diluted with
water (5 mL)
and washed with ether (10 mL). The aqueous layer was taken up in ethyl acetate
(60 mL) and
partitioned with 10% citric acid (30 mL). The organic layer was washed with
water and brine,
dried and concentrated to give a clear syrup 8c (R9=3-(4-chlorophenyl)propyl)
(105 mg, 100%).
[0203] MS(ESPOS): 390.4 [M + Na]+, 268.4 [M - Boc + H]+.
Method N
[0204] To a solution of aldehyde 8a (406.5 mg, 1.17 mmol, 1 equiv) in dimethyl
acetamide
(0.25 mL) at 0 °C was added dibromodifluoromethane (0.21 mL, 2.34 mmol,
2 equiv). To the
stirred mixture was added a solution of triphenylphosphine (0.61 g, 2.34 mmol,
2 equiv) in
dimethyl acetamide (0.5 mL) over a period of 20 minutes under nitrogen. The
reaction mixture
was warmed to rt and stirred for 30 minutes, and then was added to an
activated zinc (0.25g,
3.82 mmol, 3.3 equiv) with the aid of dimethyl acetamide (0.3 mL). The
resulting reaction
mixture was stirred at 110°C for 1 h and cooled to rt and filtered with
the aid of
dimethylacetamide (7 mL). The filtrate was poured into ice water (100 mL) and
extracted with
ether (150 mL). The ether layer was washed with brine, dried and concentrated.
The residue
54



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
was purified by chromatography to give a clear oil 8b (R9'=3,3-difluoroprop-2-
enyl) (182 mg,
41 %).
[0205] MS(ESPOS): 282.4 [M - Boc + H]+.
[0206] To a solution of 8a (R9'=3,3-difluoroprop-2-enyl) (126 mg, 0.33 mmol)
in MeOH (35
mL) was added 10% palladium on carbon (Degussa wet form 50% w/w water) (120
mg). The
reaction mixture was stirred at rt under hydrogen (1 atm) overnight and was
filtered through
celite with the aid of MeOH. The filtrate was concentrated to give a clear
syrup 8c (R9=3,3-
difluoropropyl) (97 mg, 100%).
[0207] MS(ESPOS): 194.4 [M - Boc + H]+, MS(ESNEG): 292.4 [M - H] .
Method O
[0208] [0202] To a solution of aldehyde 8b (258 mg, 0.74 moral, 1 equiv) in
THF (3 mL)
at 0°C was added tetraallyltin (178 ~.L, 0.74 mmol, 1 equiv), followed
by the drop-wise addition
of boron trifluoride etherate (94.3 wL, 0.74 mmol, 1 equiv) over a period of
15 min. The
reaction mixture was stirred at 0°C for 1.5 h. Then a solution of
potassium fluoride (125 mg) in
water (1.25 mL) was added. The resulting mixture was warmed to rt and stirred
at rt for 20 min.
This was followed by the addition of methanol (10 mL) and the resulting
mixture was stirred at
rt for another 20 min. The reaction mixture was filtered over celite. The
filtrate was evaporated
to dryness. The residue was diluted with dichloromethane (100 mL), washed with
water (50
mL), dried, concentrated and purified by chromatography to give a clear oil 9a
(R9=2-
hydroxypent-4-enyl) (261 mg, 90%): MS(ESPOS): 412.5 [M + NA]+, 290.4 [M - Boc
+ H]+.
[0209] [0203] To a solution of dimethylsulfoxide (0.17 mL, 2.42 mmol, 3 equiv)
in
dichloromethane (0.5 mL) at -72°C was added a 2 M solution of oxalyl
chloride in
dichloromethane (0.61 mL, 1.21 mmol, 1.5 equiv) over a period of 1 min. The
mixture was
stirred at -72°C for 25 min, followed by the drop-wise addition of a
solution of the alcohol 9a
(314 mg, 0.81 mmol, 1 equiv) in dichloromethane (1.4 mL) over a period of 2
min. The reaction
mixture was stirred at -72°C for 25 min, then warmed to -50°C
and stirred for an additional 2 h.
Triethylamine (0.56 mL, 4.04 mmol, 5 equiv) was added and stirred at -
50°C for 25 min. The
mixture was diluted with ethyl acetate (100 mL), washed with 5% citric acid
(100 mL), water,
saturated aqueous NaHC03 and brine, dried, evaporated and coevaporated with
anhydrous
toluene to give a clear syrup 9b (R9=2-propenylcaxboxymethyl) (287 mg, 92%).
MS(ESPOS):
288.5 [M - Boc + H]+; MS(ESNEG): 386.2 [M - H] .



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0210] To a solution of ketone 9b (225.1 mg, 0.58 mmol, 1 equiv) in
dichloromethane (2
mL) at -78°C was added diethylaminosulfur trifluoride (0.46 mL, 3.49
mL, 6 equiv). The
reaction mixture was warmed to rt and stirred at rt for 3 h, followed by an
addition of additional
(diethylamino)sulfur trifluoride (0.46 mL, 3.49 mL, 6 equiv) at -78°C.
The mixture was warmed
to rt and stirred overnight. Then the mixture was diluted with dichloromethane
(60 mL), washed
with sat. aqueous NaHC03 (1 x), brine (1 x), dried, and evaporated. The
residue was purified by
chromatography to give a yellow oil 9c (X, X=fluoro, flouro) (75 mg, 32%).
[0211] MS(ESPOS): 310.5 [M - Boc + H]+.
[0212] To a solution of 9c (R9=2,2-difluoropent-4-enyl) (85 mg, 0.21 mmol) in
MeOH (20
mL) was added 10% palladium on carbon (Degussa wet form 50% w/w water) (100
mg). The
reaction mixture was stirred at rt under hydrogen (1 atm) overnight, was
filtered through celite
with the aid of MeOH (10 mL). To the filtrate was added 10% palladium on
carbon (Degussa
wet form 50% w/w water) (130 mg). The reaction mixture was stirred at rt under
hydrogen (1
atm) overnight, was filtered through celite with the aid of MeOH (10 mL). The
filtrate was
concentrated to give haloalkyl N-Boc-amino acid 9d (X,X=fluoro, fluoro) (67.7
mg, 100%) as a
clear syrup.
[0213] MS(ESPOS): 344.4 [M + Na]+, 222.4 [M - Boc + H]+ MS(ESNEG): 320.2 [M -
H] .
Method P
[0214] To 4-propylpyridine (2.5 g, 20 mmol), 30% hydrogen peroxide (2.4 g) was
added and
refluxed overnight. The solvent was removed and the resulting residue was
taken in DCM (30
mL). Trimethylsilyl cyanide (2.6 g, 26 mmol) was added to the above solution
followed by
dimethylcarbamyl chloride (2.8 g, 26 mmol), and the reaction mixture was
stirred at rt
overnight. Potassium carbonate (10%, 100 mL) was added. The organic layer was
separated,
dried over sodium sulfate and then concentrated to obtain 4-propyl-2-
cyanopyridine (2.5 g,
93%). It was then refluxed in hydrochloric acid (6N, 60 mL) for overnight. The
4-propyl-2-
carboxylic acid pyridine lOb (R9=propyl) was obtained after crystallization
from acetonitrile
(2g, 71 %).
[0215] MS(ESPOS): 166 (M+H);1H NMR (300 MHa, CD30D) S 8.75 (dd, J= 9.0, 3.0,
1),
8.42 (s, 1), 8.08 (dd, J= 9.0, 3.0, 1), 3.00 (t, J= 7.5, 2), 1.82 (m, 2), 1.05
(t, J= 7.2, 3).
56



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
I
0
RZ
R3
O O N HN
/ ~ a. ~ ~ c.
-N OH HO ~"'SRS
OH b.
13a =
HO OH
13b
Rg, Rs
O O
d. ~ ~ ~ e.
-~N~., RZ r NH HN~..
13b
O O
HO ~v'SR~ HO ~"SRS
HO .~OH HO OH
13c
Method Q
[0216] A mixture of picolinic acid (Aldrich) (20 g, 162 mmol, 1 equiv) and
sodium bromide
(33.43 g, 325 mmol, 2 equiv) in thionyl chloride (81 mL) was refluxed for 5 h.
The solvent was
removed under vacuum. Absolute methanol (160 mL) was added and the mixture was
stirred at
rt for 30 minutes. The solvent was evaporated, and the residue was taken up in
5% sodium
bicarbonate and extracted with ethyl acetate (3x). The organic layers were
combined and dried
over MgS04 and evaporated. The residue was purified by chromatography to give
4-
chloropicolinic acid methyl ester as a white solid (19.9 g, 72%): 1H NMR (300
MHz, CDC13) 8
8.63 (d, J= 5.4, 1), 8.13 (d, J= 2.1, 1), 7.48 (dd, J= 2.0, 5.3, 1), 4.00 (s,
3).
[0217] A mixture of 4-chloropicolinic acid methyl ester (2.4 g, 14.1 mmol),
57% hydroiodic
acid (13.3 mL) and 50% aqueous hypophosphorous acid (0.66 mL) was stirred at
85 °C for 2 h
and then was stirred at 107 °C overnight. The mixture was cooled to 95
°C. At this temperature
over 30 minutes 10 M sodium hydroxide aqueous solution (4.2 mL) was added,
followed by the
addition of water (15.2 mL). The mixture was cooled to rt and stirred at rt
for lh. The
precipitate was filtered, washed with cold water and dried under high vacuum
overnight to give
4-iodopipecolinic acid 13a (3.5 g, 66%): 1H NMR (300 MHz, DMSO d6) 8 8.39 (d,
J= 5.1, 1),
8.35 (d, J=1.8, 1), 8.07 (dd, J=1.7, 5.2, 1); MS (ESPOS): 250.2 [M + H] +.
[0218] To a mixture of 7-Me MTL HCl salt 2b (RI=Me, R2=Me) (200 mg, 0.69 mmol,
1
equiv) in dry DMF (1.8 mL) at 0 °C was added triethylamine (0.50 mL,
3.61 mmol, 5.2 equiv),
followed by the addition of BSTFA (0.28 mL, 1.04 mmol, 1.5 equiv). The
reaction mixture was
57



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
stirred at 0 °C for 10 minutes, and then was stirred at rt for 50
minutes. To the reaction mixture
was added the acid 13a (341 mg, 0.90 mmol, 1.3 equiv) and HATU (423 mg, 1.11
mmol, 1.6
equiv). The reaction mixture was stirred at rt for 3 h. The reaction mixture
was evaporated to
dryness, taken up in ethyl acetate, washed with water (1 x), sat. NaHC03 (1 x)
and brine. The
organic layer was dried over Na2S04 and evaporated to give a yellow residue
which was
dissolved in methanol (20 mL) to which was added dry Dowex resin (250 mg). The
reaction
mixture was stirred at rt for 1 h. The resin was removed by filtration and the
crude product
eluted with 2M ammonia in methanol. The methanolic eluent was evaporated, and
the resulting
residue was purified by chromatography to provide a white solid 13b (Rl=Me,
R2= Me, R3=H)
(250 mg, 75%): iH NMR (300 MHz, CD30D) 8 8.46 (d, J=1.8, 1), 8.30 (d, J= 5.4,
1), 7.98
(dd, J= 1.8, 5.1, 1), 5.25 (d, J= 6.0, 1), 4.32-4.23(m, 2), 4.09 (dd, J= 5.7,
10.2, 1), 3.87 (d, J=
3.0, 1), 3.54 (dd, J= 3.3, 10.2, 1), 2.24-2.15 (m, 1), 2.11 (s, 3), 0.99-0.96
(m, 6); MS (ESPOS):
483.5 [M + H] +; MS (ESNEG): 481.4 [M - H]'.
[0219] To a dry flask was added 13b (Rl=Me, R2= Me, R3=H) (133.9 mg, 0.28
mmol, 1
equiv), triphenylphosphine (46.7 mg, 0.18 mmol, 0.64 equiv), copper (I) iodide
(33.9 mg, 0.18
mmol, 0.64 equiv), palladium acetate (20 mg, 0.09 mmol, 0.32 equiv) and
triethylamine (1.6
mL). The mixture was deaerated with nitrogen, followed by addition of 3-prop-2-
ynyl-
cyclopentane (120 mg, 1.11 mmol, 4 equiv). The mixture was stirred at 50 oC
overnight. The
solvent was removed under vacuum to give a dark residue. The residue was
purified by
chromatography to give 13c (Rl=Me, R9'= 3-cyclopentyl-prop-1-ynyl, R2= Me,
R3=H) as a
yellow solid (106 mg, 83%): 1H NMR (300 MHz, CD30D) 8 8.55 (d, J= 4.8, 1),
7.98 (s, 1),
7.47 (dd, J= 1.7, 5.0, 1), 5.26 (d, J= 5.4,1), 4.33-4.22 (m, 2), 4.10 (dd, J=
5.5, 10.4, 1), 3.86
(d, J= 3.3, 1), 3.55 (dd, J= 3.3, 10.5, 1), 2.49 (d, J= 6.9, 2), 2.26-2.12 (m,
2), 2.11 (s, 3), 1.93-
1.82 (m, 2), 1.73-1.55 (m, 4), 1.43-1.31 (m, 2), 1.00-0.96 (m, 6); MS (ESPOS):
463.6 [M + H] +;
MS (ESNEG): 461.5 [M - H]-.
9
a. ~ ~ ~ O b. ~ ~ ~ O c. ~ ~ ~ O
13a
-N OMe OMe OMe
14a 14b 14c
58



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Method R
[0220] To a solution of 13a prepared in general method Q (5 g, 13.26 mmol) in
methanol .
(500 mL) was added a few drops of conc. sulfuric acid. The reaction mixture
was refluxed
overnight. The solvent was evaporated and the residue was purified by
chromatography to give
4-iodopipecolinic acid methyl ester 14a as a yellow solid (3.0 g, 86%}: 1H NMR
(300 MHz,
CDC13) 8 8.49 (d, J= 1.5, 1), 8.37 (d, J= 5.4, 1), 7.85 (dd, J= 1.6, 5.2, 1),
4.00 (s, 3); MS
(ESPOS): 264.3 [M + H~ +.
[0221] To a dry flask were added 14a (1 g, 3.8 mmol, 1 equiv),
triphenylphosphine (79.7
mg, 0.3 mmol, 0.08 equiv), copper (I) iodide (57.9 mg, 0.3 mmol, 0.08 equiv),
palladium acetate
(34.1 mg, 0.15 mmol, 0.04 equiv) and triethylamine (14 mL). The mixture was
deaerated with
nitrogen, followed by addition of 3-butyn-1-of (0.53 g, 7.6 mmol, 2 equiv).
The mixture was
stirred at rt for 3 h. The solvent was removed under vacuum to give a dark
residue. The residue
was purified by chromatography to give 14b (R9'= 3-hydoxy-but-1-ynyl) as a
yellow oil (0.78 g,
100%): 1H NMR (300 MHz, CDC13) S 8.66-8.63 (m, 1), 8.09-8.08 (m, 1), 7.43-7.40
(m, 1), 3.99
(s, 3), 3.88-3.82 (m, 2), 2.72 (t, J= 6.3, 2). MS (ESPOS): 206.4 [M + H] +.
[0222] To a solution of the above 14b (R9'= 3-hydoxy-but-1-ynyl) (0.78 g, 3.8
mmol) in
methanol (40 mL) was added 10 % palladium on carbon (0.4 g). The flask
containing the
reaction mixture was purged and charged with hydrogen (1 atm) and stirred at
rt overnight. The
palladium was removed by filtration and the filtrate was concentrated to give
14c (Rg=3-
hydroxybutyl) as an oil (0.77 g, 97%): 1H NMR (300 MHz, CDC13) b 8.60 (d, J=
4.5, 1), 7.97
(d, J=1.2, 1), 7.29 (dd, J= 1.6, 5.0, 1), 3.99 (s, 3), 3.67 (t, J= 6.3, 2),
2.72 (t, J= 7.7, 2), 1.81-
1.69 (m, 2), 1.62-1.54 (m, 2); MS (ESPOS): 210.4 [M + H] +
59



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
ORIo
O 3
OH OR~o N R
R2
\ ~. HNIu~,.
N C02H N COZH
HO
15a '~UgR~
HO~
OH
OR~o
15b
O 3
N R
H R2
lii~.
d.
HO ~~
''~,q/SR1
HO ~~pH
Method S
[0223] To 4-hydroxypyridine-2-carboxylic acid (200 mg, 1.4 mmol) in DMF (2
mL),
potassium carbonate (397 mg, 2.8 mmol) was added followed by ~-bromobutane
(197 mg, 1.4
mmol), warmed at 60°C for overnight. The solvent was removed to obtain
the crude ester
product. The crude ester (360 mg, 1.4 mmol) was dissolved in THF (4 mL),
lithium hydroxide
(72 mg, 1.7 mmol) was added, and the reaction mixture stirred at room
temperature for 2 h. The
residue obtained on removal of solvent was purified by silica gel
chromatography using 10%
MeOH in DCM to provide 4-butoxypyridine-2-carboxylic acid 15a (Rl°=
butyl) (100 mg, 43%).
iH NMR (300 MHz, CD30D) 8 8.37 (d, J--6.0, 1), 7.63 (d, J--2.7, 1), 7.07 (dd,
J--2.7, 6.0, 1),
4.15 (t, J--6.6, 2), 1.82 (m, 2), 1.54 (m, 2), 1.01 (t, J--7.5, 3). MS (ES-):
194 (M-1).
[0224] To 4-butoxypyridine-2-carboxylic acid 15a (Rl°=Butyl) (100 mg,
0.5 mmol) in DMF
(2 mL), 7-methyl a-thiolincosaminide 2b (Rl=Me, Ra=Me) (147 mg, 0.5 mmol) was
added,
followed by HBTU (214 mg, 0.55 mmol) and DIEA (132 mg, 1 mmol). The reaction
mixture
was stirred at rt for 2 h. Then solvent.was removed. Purification of the crude
material was
carried out by silica gel column chromatography to obtain compound 15b (Rl=Me,
R2=Me,
R3=H, Rl°=butyl) (201 mg, 91%):



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0225] 1H NMR (300 MHz, CD30D) 8 8.42 (m, 1), 7.96 (s, 1), 7.09 (m, 1), 5.27
(d, J--5.4,
1), 4.10-4.87 (m, 3), 3.85 (d, J--3.3, 1), 3.76 (m, 1), 2.11 (m, 4), 1.81 (m,
2), 1.49 (m, 4), 0.99
(m, 9). MS (METHOD ES+): 428 (M+1).
[0226] To a solution of the pyridine 15b (Rl=Me, R2=Me, R3=I-I,RI°=
butyl) (200 mg, 0.46
mmol) in water (10 mL), AcOH (3 mL) and MeOH (2 mL), was added Pt02 (200 mg)
and the
resulting reaction mixture shaken under 55 psi hydrogen overnight. Residual
catalyst was
removed by filtration through celite, and the solvent was removed to obtain
the crude product.
Purification was carried out by silica gel column chromatography using 20%
MeOH in DCM to
obtain lincosamide analog 1 (Rl=Me, R2=Me, R3=H, Rl° = butyl) (12 mg,
6%).1H NMR (300
MHz, CD30D) 8 5.25 (d, J--5.4, 1), 4.22 (dd, J--10.2; 3.3, 1), 4.08 (m, 2),
3.81 (d, J--3.0, 1), 3.70
(m, 1), 3.54 (m, 4), 3.43 (m, 2), 2.90 (m, 1), 2.41 (m, 1), 2.19 (m, 1), 2.10
(s, 3) 1.45 (m, 6), 0.92
(m, 9); MS (ES+): 435 (M+1).
Example 1
Preparation of 1-(4-ethylpiperid-6-yl)-N {1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl{acetamide
'O
H HH,HH, ~.
H ~~~SMe
HOr~~H
[0227] 4-Ethyl pyridine-2-carboxylic acid HCl salt (Toronto) (117 mg, 0.64
mmol) was
suspended in dry acetonitrile (4 mL). Triethylamine (180 pL, 1.28 mmol) was
added and the
reaction mixture was cooled to 0°C. Isobutyl chloroformate (129 ~.L,
0.62 mmol) was added
and the reaction mixture was warmed to 4°C. After 1.5 h the activated
ester solution was
transferred to a solution of 2b (Ri=Me, R2=Me), prepared as in Method C, in
1:1 acetone/water
(2 mL) and warmed to 30°C to dissolve. Triethylamine (80 ~L, 0.057
mmol) was then added to
the reaction mixture. The reaction mixture was stirred for 10 h at rt, then
evaporated to dryness
and chromatographed on silica 94:5 dichloromethane:0.25% ammonia in methanol
to provide
llb (Rl=Me, Ra=Me R3=H, R9=ethyl) (167 mg 69.7%).
MS (ESPOS): 385.2 [M+H].
61



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0228] A solution of pyridine llb (m=2, Ri=Me, R2=Me R3=H, R9=ethyl) (167 mg,
0.435
mmol) in 3:2 methanol/water (20 mL) was added to platinum(IV)oxide (339 mg,
0.521 mmol) in
a Parr bottle. Concentrated HCl (52 ~.L, 0.52 mmol) was then added. The bottle
was purged,
and charged with H2 to 65 psi and shaken for 24 h. The reaction mixture was
filtered through
celite and rinsed with methanol. The combined filtrate was evaporated to
dryness and
chromatographed on silica 88:12 to 80:20 dichloromethane: 0.25% ammoniain
methanol to give
43 mg of a high Rf product and 49 mg of a mixed fraction. Chromatography of
the low Rf
fraction on fluorosil 84:16 to 80:20 dichloromethane: 0.25% ammoniain methanol
provided 1-
(6-(S~-4-(R)-ethylpiperid-6-yl)-N f 1-(R)- [2-(S)-3-(S),4-(S),5-(R)-trihydroxy-
6-(R)-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (21.9 mg,
12.9%), which was
taken up in 1:1 acetonitrile:water (50 mL), 0.2~, millipore filtered, and
cooled to 0°C. 1N HCl
(67 ~,L) in water (20 mL) was added and re-lyophilized to provide the HCl salt
(24.0 mg) as a
colorless powder.
[0229] 1H NMR (300 MHz, D2O) 8 5.32 (d, J= 5.8, 1), 4.14-4.06 (m, 1), 4.12 (s,
2), 3.85 (d,
J= 3.30, 1), 3.60 (dd, J= 3.3, 10.4, 1), 3.30 (dd, J= 2.5, 11.8, 1), 3.09 (m,
1), 2.56 (ddd, J= 2.8,
12.9, 15.7, 1), 2.14 (s, 3), 2.14-2.05 (m, 1), 1.96-1.90 (m, 1), 1.74-1.69 (m,
1), 1.45-1.35 (m, 1),
1.33-1.23 (m, 2), 1.08-0.98 (m, 2), 0.86 (m, 9); MS(ESPOS)391.4 [M+H], 803.5.4
[2M+Na],
(ESNEG): 389.5 [M-H].
Example 2
Preparation of 1-(-4-n-propyl-N methylpyrrolidin-2-yl)-N f 1- [3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
~i,.
~: ~~O
Hf~
H ~~ 6Me
HQr~~H
[0230] 4-n-Propylhygric acid prepared by the method of Hoeksema, H. et. al.
Journal of the
American Chemical Society, 1967, 89 2448-2452 (157 mg, 0.76 mmol) was
suspended in dry
acetonitrile (5 mL). Triethylamine (421 ~.L, 3.02 mmol) was added and the
reaction mixture
was cooled to 0°C. Isobutyl chloroformate (98 ~.L, 0.76 mmol) was added
and after 10 min the
reaction was allowed to warm to 4°C. After 1.5 h a solution of 2b
(Ri=Me, R2=Me), from
62



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Method C (190 mg, 0.76 mmol) in 1:1 acetone:water (5 mL) was added and the
reaction mixture
was stirred for l Oh at rt. The reaction mixture was evaporated to dryness and
chromatographed
on silica 94:6 dichloromethane:0.25% ammonia in methanol. Fractions 14-18
contained the
product as a colorless oil (50.2 mg, 16.5%).
[0231] 1H NMR (300 MHz, Da0) b 5.33 (d, J= 6.0, 1), 4.27-4.22 (m, 1), 4.18 (s,
1), 4.09
(dd, J = 5 . 8, 10.2, 1 ), 3 .92-3 . 81 (m, 1 ), 3 .92-3 .81 (m, 1 ), 3 .64-3
. 5 9 (m, 1 ), 2.92 (s, 3 ), 2.92-2. 8 5
(m, 1), 2.35-2.28 (m, 3), 2.13 (s, 3), 1.46-1.41 (m, 2), 1.40-1.28 (m, 2),
0.89-0.84 (m, 9);
MS(ESPOS): 405.5 [M+H].
Example 3
Preparation of 1-(-4-n-propyl-N-methylpyrrolidin-2-yl)-N-{1- [3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-methyl-3-cyanoprop-1-yl] acetamide
~6Me
[0232] Lincosamine 2b (Rl=Me, R2=CHaCN) (54.2 mg, 0.20 mmol) prepared by
Method E
was dissolved in DMF (0.7 mL). The reaction mixture was cooled to 0°C
and triethylamine
(170 ~L, 1.2 mmol) and BSTFA (96 ~L, 0.36 mmol) was added. The reaction
mixture was
allowed to warm to rt, and stirred at rt for 1 h. 4-v~-Propylhygric acid
prepared by the method of
Hoeksema, et al., J. Am. Chem. Soc., 1967, ~9 2448-2452 (66.4 mg, 0.32 mmol)
and HATU
(149 mg, 0.39 mmol) were added, and the mixture was stirred at rt for 3 h. DMF
was removed
and the residue was dissolved in DCM (100 mL), washed with saturated NaHC03
(30 mL) and
brine (30 mL), and dried over sodium sulfate. The residue obtained by removing
the solvent
was dissolved in methanol (20 mL) and treated with Dowex resin H+ (300 mg) for
15 min. The
crude product was eluted from the resin by washing with 5% TEA in MeOH (25 mL
x 15 min x
2) and 5% TEA in MeGN (25 mL x 15 min). The combined eluent was evaporated to
dryness
and purified by silica gel column chromatography using 7% 0.25M NH3 in
methanol in
dichloromethane as the eluent (24 mg, 28°/°).
63



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0233] 1H NMR (300 MHz, D20) ~ 5.61 (d, J= 5.8, 1), 4.59 (d, J=10.2, 1), 4.46
(d, J=
10.2, 1), 4.46 (dd, J= 6.0, 10.4, 1), 4.05 (d, J= 3.0, 1), 3.84 (dd, J= 3.3,
10.4, 1), 3.48 (dd, J=
5.8, 8.0, 1), 3.34 (dd, J= 5.2, 10.2, 1) 2.81-2.61 (m, 2), 2.65 (s, 3), 2.43
(s, 3), 2.31-2.10 (m, 2),
1.32 (d, J= 6.0; 1), 1.18 (t, J=7.1, 3); MS(ESPOS): 430.5 [M+H] MS(ESPOS):
428.5 [M-H].
Example 4
Preparation of 1-(-4-ethylpiperidyl)-N-{1- [3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-hydroxy-2-methylprop-1-yl)acetamide
OH
INi H~(N, ~.
H ~~~SMe
HOr~OH
[0234] Lincosamine 3b (Rl=Me, R2=Me) prepared by Method F (54.2 mg, 0.20 mmol)
was
dissolved in DMF (1.0 mL). The reaction mixture was cooled to 0°C and
triethylamine (178 ~,L,
1.3 mmol) and BSTFA (85 ~L, 0.32 mmol) were added. The reaction mixture was
allowed to
warm to rt, and stirred at for 1 h. 4-Ethyl pyridine-2-carboxylic acid HCl
salt (Toronto) (55.3
mg, 0.29 mmol) and HATLT (122 mg, 0.32 mmol) were added, and the mixture was
stirred at rt
for 3 h. DMF was removed and the residue was dissolved in THF (10 mL), and
treated with
(600 mg) Amberlite A-26 F-form resin and catalytic TBAF for Sh. The crude
product was
obtained by removal of the resin and evaporation of the solvent to dryness and
purified by silica
gel column chromatography using 10% 0.25M NH3 in methanol in dichloromethane
as the
eluent to provide the pyridine product llb (m=2, RI=Me, R2=Me R3=OH, R9=ethyl)
(26 mg,
33%).
[0235] MS(ESNEG): 399.5 [M-H].
[0236] A solution of pyridine llb (m=2, Rl=Me, R2=Me R3=OH, R9=ethyl) (26 mg,
0.065
mmol) in 3:2 methanol:water (10 mL) was added to platinum(IV)oxide (51 mg) in
a Parr bottle.
Concentrated HCl (6.0 ~L, 0.072 mmol) was then added. The bottle was purged
and charged
with HZ to 65 psi and shaken for 24 h. The reaction mixture was filtered
through celite and
rinsed with methanol. The combined filtrate was evaporated to dryness and
chromatographed
64



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
on silica 80:20 dichloromethane: 0.25% ammonia in methanol to give a high Rf
product and the
title compound (5.8 mg, 21.8%).
[0237] iH NMR (300 MHz, D20) 8 5.37 (d, J= 6.0, 1), 4.41 (d, J= 9.6, 1), 4.32
(d, J= 9.3,
1), 4.08 (dd, J= 6.6, 11.0, 1), 3.93-3.90 (m, 2), 3.59 (dd, J= 3.0, 10.7, 1),
3.93-3.90 (m, 2), 3.04
(apt dt, J= 7.1, 14.6, 14.6, 1), 2.24-2.18 (m, 1), 2.20 (s, 3), 1.70-1.60 (m,
1), 1.42-1.13 (m, 1),
0.88 (dd, J=6.0, 7.4, 1); MS(ESPOS): 407.4 [M+H].
Example 5
Preparation of 1-(-4-n-propyl-N methylpyrrolidin-2-yl)-N ~1- [3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-hydroxyiminoprop-1-yl] acetamide
W ,.
O
i H~" 'NOH
H ~~~SMe
HO ~Z7 H
[0238] Triethylamine (0.041 mL, 0.28 mmol) and BSTFA (0.24 mL, 0.94 mmol) were
added to the crude oxime 4b (Rl=Me, R7=H) prepared by Method G (50 mg, 0.19
mmol) in
DMF (3 mL) at 0°C and the mixture was stirred at rt overnight. Next, 4-
h-propylhygric acid (63
mg, 0.37 mmol) and HATU (142 mg, 0.37 mmol) were added and the mixture was
stirred at rt
for 4 h. DMF was removed and the residue was extracted with dichloromethane
(100 mL) and
washed with saturated bicarbonate (20 mL) and brine (20 mL). The residue
obtained on removal
of dichloromethane was then treated with 10% TFA in dichloroethane (10 mL) and
dimethyl
sulfide (0.5 mL) for 1 h. The solvent was then removed to obtain the crude
product, which was
purified by silica gel column chromatography using 20% methanol in
dichloromethane as the
eluent to provide the title compound (20 mg, 25%).
[0239] TLC: Rf= 0.67 (20% methanol in dichloromethane);1H NMR (300 MHz, CD30D)
8
0.93 (t, J= 6.8, 3), 1.31-1.44 (m, 4), 1.88 (s, 3), 1.99 (s, 3), 2.09 (m, 2),
2.11 (m, 1), 2.62-2.98
(m, 3), 2.76 (s, 3), 3.60 (m, 2), 4.10 (dd, J= 5.7, 10.20, 1), 4.27 (d, J=
9.6, 1), 5.23 (d, J= 5.5,
1), MS(ESPOS): 420 (M+H).



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 6
Preparation of 1-(-4-n-propyl-N methylpyrrolidin-2-yl)-N {1- [3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-methoxyiminoprop-1-yl) acetamide
~i,
C,~~'~O
Hf~
~6Me
[0240] From crude oxime product 4b prepared by Method H (R1=Me, R7=Me), the
title
compound was prepared as in Example 5 (10 mg, 47%).
[0241] TLC: Rf= 0.55 (10% methanol in dichloromethane); 1H NMR (300 MHz,
CD30D) 8
0.91 (m, 3), 1.32 (m, 4), 1.88 (s, 3), 1.98 (s, 3), 1.78-2.04 (m, 2), 2.34 (s,
3), 2.90 (dd, J= 5.1,
6.30, 8.10 1), 3.21 (dd, J= 6.3, 10.2, 1), 3.57 (dd, J= 3.3, 10.2, 1), 4.23
(dd, J= 5.4, 10.2, 1),
5.25 (d, J= 5.7, 1); MS(ESPOS): 434 (M+H).
Example 7
Preparation of 1-(-3-n-butylpiperid-6-yl)-N ~1- [3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
~O
H HN
H ~~ ~SMe
HOr~H
[0242] Lincosamine intermediate 2b (Ri=Me, Ra=Me), prepared by Method C, was
dissolved in DMF (2 mL). Triethylamine (80 mg, 1 mmol) and BSTFA (307 mg, 1.1
mmol)
were added, and the mixture was stirred at rt for 1.5 h. Next, fusaric acid
(143 mg, 0.7 mmol)
and HATU (184 mg, 0.5 mmol) were added, and the mixture was stirred at rt for
3 h. DMF was
removed and the residue was dissolved in EtOAc (50 mL), washed with sodium
bicarbonate
(10%, 30 mL) and brine (30 mL), and dried over sodium sulfate. The residue
obtained by
removing the solvent was dissolved in methanol and treated with Dowex resin H+
for 1 h. The
crude product obtained by filtering the resin and removing the solvent was
purified on silica gel
66



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
column chromatography using 10% methanol in dichloromethane as the eluent to
give the title
compound (100 mg, 61 %).
[0243] TLC Rf= 0.6 (10% methanol in dichloromethane);1H NMR (300 MHz, CD3OD) 8
8.47 (s, 1), 8.02 (d, J= 8.1, 1), 7.80 (d, J= 8.1, 1), 5.27 (d, J= 5.4, 1),
4.31 (m, 2), 4.12 (dd, J=
5.7, 4.2, 1), 3.85 (d, J= 3.0, 1), 3.56 (dd, J= 3.3, 6.9, 1), 2.80 (m, 2),
2.24 (m, 1), 2.11 (s, 3),
1.67 (m, 2),1.41 (m, 2), 1.00 (m, 9); MS(ESPOS): 413 (M+H).
[0244] Pt02 (50 mg, 0.22 mmol) was added to compound llb (m=2, Rl=Me, Ra=Me,
R3=H,
R9=butyl), (70 mg, 0.16 mmol) in methanol (2 mL), water (l OmL), and acetic
acid (3 mL), and
the mixture was hydrogenated at 50 psi overnight. The product obtained after
filtering the
catalyst and removing the solvent was purified on silica gel column
chromatography using 30%
methanol in dichloromethane as the eluent (16 mg, 46%).
[0245] TLC Rf= 0.7 (30% methanol in dichloromethane);1H NMR (300 MHz, CD30D) ~
5.24 (d, J= 5.7, 1), 4.16 (m, 3), 3.82 (d, J= 3.3, 1), 3.53 (m, 2), 2.93 (m,
2), 2.09 (s, 3), 1.93 (m,
1), 1.76 (m, 2), 1.50 (m, 1), 1.30 (m, 7), 0.92 (m, 9); MS(ESPOS): 419 (M+H).
Example 8
Preparation of 1-(4-(R,S)-n-pentylpyrrolidin-2-yl)-N ~1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide
~~O
H~~,.
H ~~iSMe
HO ~~OH
[0246] Triethylamine (0.2 mL, 1.44 mmol, 3.6 equiv), followed by BSTFA (0.2
mL, 0.76
mmol, 1.9 equiv), were added to a stirred suspension of 2b (Rl=Me, Ra=Me)
prepaxed by
Method C (100.4 mg, 0.4 mmol, 1 equiv) in anhydrous DMF (2 mL) at 0°C
and under nitrogen.
The resulting mixture was stirred at 0°C for 10 min, and then at rt for
50 min. The resulting
solution was cooled to 0°C and a solution of 6c (R9~entyl) (Scheme 6)
prepared as described in
Birkenmeyer, R. D; et al; Journal of Medicinal Chemistry 1972, I5, 1255-1259.
(144 mg, 0.51
mmol, 1.2 equiv) in anhydrous DMF (1.5 mL) was added, followed by solid HATU.
The
reaction mixture was allowed to warm to rt, and after 2 h, the reaction
solution was evaporated
to dryness under vacuum. The residual oil obtained was diluted with EtOAc (150
mL), washed
67



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
sequentially with 10% citric acid (2 x 30 mL),1:1 saturated aqueous NaHC03,
water (2 x 30
mL), and brine (30 mL), dried over Na2S04, and evaporated to dryness.
(0247] 1,2-Dichloroethane (8 mL), followed by dimethyl sulfide (180.3 ~l), TFA
(2.7 mL),
and water (180.3 ~l) were added to the crude product (267.5 mg) obtained
above. The resulting
mixture was stirred at rt for lh and evaporated to a minimal volume, diluted
with DCE (3-x 30
mL}, and evaporated to dryness. The residue obtained was purified by
chromatography over
silica gel, with a gradient eluent of 8-10% methanol ammonia in
dichloromethane. The desired
fractions were pooled together, evaporated to dryness, and lyophilized to
furnish the title
compound as a white fluffy powder (35.6 mg, 21.2%).
[0248] TLC, Rf= 0.15 (16% 0.25M methanolic ammonia in.dichloromethane ). 1H
NMR
(300MHz, Da0) ~ 5.4 (d, J= 5.8, 1), 3.91(s, 1), 3.69-3.66 (m, 3), 2.1 (s, 3),
1.32-1.15 (m, 3.37),
0.93. 0.87 (m, 9.8); MS(ESPOS): 419.5 [M+H], (ESNEG): 417.45 [M-H].
Example 9
Preparation of 1-[4- (3-methylbut-1-yl)pyrrolidin-2-yl]-N f 1- [3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl) acetamide
~~O
INi HN,~.
H ~~~6Me
HO~~H
(0249] Triethylamine (0.13 mL, 0.96 mmol, 3.2 equiv), followed by BSTFA (0.12
mL, 0.45
mmol, 1.5 equiv), were added to a solution of 2b (Rl=Me, Ra=Me) prepared by
Method C (75
mg, 0.30 mmol, 1 equiv) in dry DMF (0.8 mL) at 0°C. The reaction
mixture was stirred at 0°C
for 10 min, and then at rt for 50 min. To the reaction mixture was added acid
7d (R9=2-
methylbutyl), prepared by method J (160 mg, 0.56 mmol, 1.9 equiv), in a 25 mL
round-bottom
flask. Then HATU was added (227 mg, 0.60 mmol, 2 equiv). The reaction mixture
was stirred
at rt for 3 h. The reaction mixture was evaporated to dryness, taken up in
ethyl acetate (100
mL), washed with 10% citric acid (2 x 60 mL), water (60 mL), half sat. NaHC03
(60 mL), and
brine. The organic layer was dried over NaaSO4 and evaporated to give a yellow
syrup.
68



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0250] Trifluoroacetic acid (5 mL) and water (0.33 mL) were added to a
solution of the
above syrup in dichloromethane (1 S mL) with methyl sulfide (0.33 mL). The
reaction mixture
was stirred at rt for 1 h. The solvent was removed under vacuum and co-
evaporated with
toluene twice. The residue was purified by chromatography to provide the title
compound (75
mg, 60%) as a white solid.
[0251] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J= 5.4, 1), 4.18-3.99 (m, 4), 3.75
(d, J= 2.4,
1), 3.51 (dd, J= 3.3, 10.5, 1), 3.38-3.31 (m, 1), 2.68 (dd, J= 8.2, 10.6, 1),
2.23-2:05 (m, 3), 2.10
(s, 3), 1.97-1.87 (m, 1), 1.59-1.47 (m, 1), 1.46-1.34 (m, 2), 1.25-1.16 (m,
2), 0.92-0.88 (m, 12).
MS(ESPOS): 419.5 [M + H]+, MS(ESNEG): 417.5 [M - H]-.
Example 10
Preparation of 1-(-4-n-pentylpyrrolidin-2-yl)-N ~1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide
'' O
H (~
H ~~6Me
HOr~ H
[0252] Triethylamine (0.6 mL, 4.33 mmol, 3.6 equiv), followed by BSTFA (0.6
mL, 2.27
mmol, 1.9 equiv), were added to a stirred suspension of 2b (Rl=Me, R2=Me)
prepared by
Method C (298.8 mg, 1.19 mmol, 1 equiv) in anhydrous DMF (5 mL) at 0°C
and under nitrogen.
The resulting mixture was stirred at 0°C for 10 min, and then at rt for
50 min. The resulting
solution was cooled to 0°C and a solution of 7d (R9=~z-pentyl) was
prepared by Method I~ (400.1
mg, 1.40 mmol, 1.2 equiv) in anhydrous DMF (5 mL) was added, followed by solid
HATU
(678.7 mg, 1.79 mmol, 1.5 equiv). The reaction mixture was allowed to warm to
rt and after 2h
the reaction solution was evaporated to dryness under vacuum. The residual oil
obtained was
diluted with EtOAc (400 mL), washed sequentially with 10% citric acid (2 x 100
mL), 1:1
saturated aqueous NaHCO3, water (2 x 100 mL), and brine (100 mL), dried over
Na2SO4, and
evaporated to dryness.
[0253] 1,2-Dichloroethane (35 mL), followed by dimethylsulfide (768 ~L), TFA
(11.5 mL),
and water (768 ~L) were added to the crude product (1.14 g) obtained above.
The resulting
mixture was stirred at rt for lh, evaporated to a minimal volume, diluted with
DCE (3 x 90 mL),
69



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
and evaporated to dryness. One-third of the residue obtained was purified by
chromatography
over silica gel, with a gradient eluent of 8-12% methanol ammonia in
dichloromethane. The
desired fractions were pooled together, evaporated to dryness, treated with
deuterium
oxide/anhydrous acetonitrile, and lyophilized to furnish a white fluffy powder
(68.2mg, 41.1%);
TLC, Rf= 0.15 ~ 16% 0.25M methanolic ammonia in dichloromethane~. 1H NMR (300
MHz,
Da0) 8 5.41 (d, J= 5.8, 1H), 4.55 (m, 1), 4.24 (s, 2), 4.14 (m, 1), 3.91(d, J--
3.3, 1), 3.70-3.66 (m,
2), 3.15 (m,l), 2.36-2.27 (m, 2), 2.19 (s, 5), 1.59-1.13 (m,9H), 0.93-0.88 (m,
9);13C NMR
(DaO,): 8 170.4, 119.4, 88.4, 70.9, 69.3, 68.8, 68.2, 60.0, 53.4, 5.1.4, 37.3,
36.7, 31.3, 27.9, 27.2,
22.3, 20.1, 14.8, 13.7, 13.3; MS(ESPOS):419.6[M+H];(ESNEG):417.5[M-H].
Example 11
Preparation of 1-(-4-n-propyl-N methylpyrrolidin-2-yl)-N (1-[3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2,2-difluoroprop-1-yl~acetamide
~s,, p F
C' ~~--r~ F
Ii Ni , .
H ~ ~ 6Me
HOr~H
[0254] 30% Trifluoroacetic acid in dichloroethane (10 mL) and dimethylsulfide
(0.5 mL)
were added to lincosamine intermediate 5b (Rl=Me), prepared by Method H (100
mg, 0.20
mmol). . The mixture was stirred at rt for 1 h. The solvent was removed and
the residue was kept
under high vacuum for 1 h. N-Methyl-4-trays-propylproline (53 mg, 0.4 mmol)
and HATU
(114 mg, 0.30 mmol) were added to the dried compound in DMF (3 mL), and the
mixture was
stirred at rt overnight. DMF was removed and the residue obtained was then
extracted with
ethyl acetate (100 mL) and washed with saturated bicarbonate (50 mL). The
organic portion
was then dried using magnesium sulfate and the solvent was removed to obtain
the crude
product. The crude product was purified on silica gel column using ethyl
acetate as the eluent
(50 mg, 46%). The product (50 mg, 0.09 mmol) was then taken in methanol (2 mL)
and water
(1 mL), to which solid potassium carbonate (124 mg, 0.90 mmol) was added and
the mixture



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
was stirred at rt for 24 h. Solvents were then removed and the crude product
was purified on
silica gel column using 20% methanol in dichloromethane as the eluent (20 mg,
52%).
[0255] TLC: Rf= 0.57 (20% methanol in dichloromethane); MS(ESPOS): 427 (M+H).
1H
NMR (300 MHz, CD30D) S 0.91 (m, 3), 1.34 (m, 4), 1.69 (t, J=19.8, 3), 1.98 (s,
3), 2.20 (m,
2), 2.46 (s, 3), 3.18 (dd, J= 5.1, 10.20, 1), 3.93 (d, J= 3.0, 1), 4.08 (dd,
J= 3.3, 10.20, 1), 4.40-
4.70 (m, 2), 5.28 (d, J= 5.4, 1).
Example 12
Preparation of 1-(-4-n-pentylpyrrolidin-2-yl)-N {1-[3,4,5-trihydroaty-6
(methylthio)tetrahydropyran-2-yl]-2,2-difluoroprop-1-yl) acetamide
O F
F
Hf~
H
H ~~6Me
HO~~H
[0256] Boc 4-t~a~s-Pentylproline 7d (R9=n-pentyl) (179 mg, 0.631 mmol), HATU
(299 mg,
0.789 mmol), and diethylisopropylamine (182 mg, 1.2 mmol) were added to
lincosamine
intermediate 5b (Rl=Me) prepared by Method H (210 mg, 0.526 mmol) in DMF (3
mL) at 0°C.
The mixture was stirred at rt overnight. After removing DMF, the residue was
taken in ethyl
acetate and washed with saturated bicarbonate (30 mL). The organic portion was
then dried
over sodium sulfate and.the solvent was removed to obtain the crude product.
The crude
product was purified by column chromatography using 30% ethyl acetate in
hexanes as the
eluent (200 mg, 57%). Potassium carbonate (450 mg, 3.0 mmol) was added to the
product (200
mg, 0.30 mmol) of the above reaction in methanol (3 mL) and water (1 mL), and
the mixture
was stirred at rt for 2 h. The solvent was removed and the residue obtained
was taken in 30%
trifluoroacetic acid in dichloroethane (10 mL) and dimethyl sulfide (0.5 mL)
and stirred for 1 h.
After removing the solvent, the crude product obtained was purified by column
using 10%
methanol in dichloromethane as the eluent (10 mg, 90%).
[0257] TLC: Rf= 0.56 (20% methanol in dichloromethane); 1H NMR (300 MHz,
CD30D) 8
0.90 (m, 3), 1.31 (m, 7), 1.44 (m, 20), 1.73 (t, J=19.5, 3), 2.02 (m, 1), 2.08
(s, 3), 2.24 (m, 2),
2.89 (t, J= 9.9, 1), 3.56 (m, 2), 3.86 (s, 1), 4.07 (dd, J= 6.0, 9.6, 1), 4.37
(m, 2), 4.63 (m, 1),
5.28 (d, J= 5.4, 1); MS(ESPOS): 441 (M+H).
71



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 13
Preparation of 1-(-4-(3-p-fluorophenyl)prop-1-ylpyrrolidin-2-yl)-N ~1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
''~ O
H(~,.
H ~ ~ ~SMe
HO~~H
[0258] Triethylamine (0.13 mL, 0.96 mmol, 3.2 equiv), followed by BSTFA (0.12
mL, 0.45
mmol,1.5 equiv), were added to a solution of 2b (Rl=Me, R2=Me) prepared by
Method C (75
mg, 0.30 mmol, 1 equiv) in dry DMF (0.8 mL) at 0 °C. The reaction
mixture was stirred at 0 °C
for 10 min, and then at rt for 50 min. The reaction mixture was added to acid
8c (R9=3-(4-
fluorophenyl)propyl) prepared by Method L (120 mg, 0.34 mmol, 1.1 equiv) in a
25 mL round
bottom flask, followed by HATU (160 mg, 0.42 mmol, 1.4 equiv). The reaction
mixture was
stirred at rt for 3 h. The reaction mixture was evaporated to dryness, taken
up in ethyl acetate
(100 mL), and washed with 10% citric acid (2 x 60 mL), water (60 mL), half
sat. NaHC03 (60
mL), and brine. The organic layer was dried over Na2S04 and evaporated to give
a yellow
syrup.
[0259] Trifluoroacetic acid (5 mL) and water (0.33 mL) were added to a
solution of the
above syrup in dichloromethane (15 mL) with methyl sulfide (0.33 mL). The
reaction mixture
was stirred at rt for 1 h. The solvent was removed under vacuum and co-
evaporated with
toluene twice. The residue was purified by chromatography to provide 1-(2-(S)-
4-(R)-(3 p-
fluorophenyl)prop-1-ylpyrrolidin-2-yl)-N {1-(S)-[2-(S)-3-(S), 4-(S), 5-(R)-
trihydroxy-6-(R)-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (90 mg, 62%) as
a white solid.
[0260] 1H NMR (300 MHz, CDC13) S 7.94 (brs, 1), 7.11-7.06 (m, 2), 6.97-6.90
(m, 2), 5.31
(d, J= 5.4, 1), 4.10 (dd, J= 5.7, 9.9, 1), 3.96-3.82 (m, 3), 3.68-3.52 (m, 2),
3.10-3.20 (m, 1),
2.70-2.60 (m, 1), 2.56 (dd, J= 7.4, 7.4, 2), 2.36-2.24 (m, 1), 2.13 (s, 3),
2.10-1.93 (m, 2), 1.85-
1.73 (m, 1), 1.64-1.50 (m, 2), 1.40-1.30 (m, 2), 0.92-0.85 °(m, 6).
MS(ESPOS): 485.5 [M + H]+,
MS(ESNEG): 483.5 [M - H]-.
72



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 14
Preparation of 1-[2-(S)-4-(R)-(3,3-difluoroprop-1-yl)pyrrolidin-2-yl]-N {1-
[3,4,5-
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
F
~\ ~O
H~ ~.
H ~~6Me
HO~~H
[0261] Triethylamine (0.13 mL, 0.96 mmol, 3.2 equiv), followed by BSTFA (0.12
mL, 0.45
mmol, 1.5 equiv), were added to a solution of 2b (Rl=Me, Ra=Me) prepared by
Method C (75
mg, 0.30 mmol, l equiv) in dry DMF (0.8 mL) at 0 °C. The reaction
mixture was stirred at 0°C
for 10 min, and then at rt for 50 min. The reaction mixture was added to acid
8c (R9=3,3-
difluoropropyl) prepared by Method N (97 mg, 0.33 mmol, 1.1 equiv) in a 25 mL
round bottom
flask, followed by HATU (170 mg, 0.45 mmol, 1.5 equiv). The reaction mixture
was stirred at
rt for 3 h. The reaction mixture was evaporated to dryness, taken up in ethyl
acetate (100 mL),
and washed with 10% citric acid (2 x 60 mL), water (60 mL), half sat. NaHC03
(60 mL), and
brine. The organic layer was dried over NaaS04 and evaporated to give a yellow
syrup.
[0262] Trifluoroacetic acid (5 mL) and water (0.33 mL) were added to a
solution of the
above syrup in dichloromethane (15 mL) with methyl sulfide (0.33 mL). The
reaction mixture
was stirred at rt for 1 h. The solvent was removed under vacuum and co-
evaporated with
toluene twice. The residue was purified by chromatography to provide 1-[2-(S)-
4-(R)-(3,3-
difluoroprop-1-yl)pyrrolidin-2-yl]-N {1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-
2-methylprop-1-yl)acetamide (81 mg, 64%) as a white solid.
[0263] 1H NMR (300 MHz, CDC13) 8 7.92 (d, J= 8.1, 1), 5.80 (dddd, J= 4.2, 4.2,
57, 57, 1),
5.31 (d, J= 5.7, 1), 4.11 (dd, J= 5.4, 9.9, 1), 3.96-3.82 (m, 3), 3.64-3.52
(m, 2), 3.23-3.10 (m,
1), 2.73-2.60 (m, 1), 2.36-2.23 (m, 1), 2.13 (s, 3), 2.18-1.95 (m, 2), 1.90-
1.73 (m, 3), 1.56-1.43
(m, 2), 0.93-0.85 (m, 6). MS(ESPOS): 427.5 [M + H]+, MS(ESNEG): 425.5 [M - H]-
.
73



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 15
Preparation of 1-(-4-(3 F-chlorophenyl)prop-1-ylpyrrolidin-2-yl)-N (1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
CI
H H~,.
O
H ~~~SMe
HO 'OH
[0264] Triethylamine (88.3 wL, 0.64 mmol, 3.2 equiv), followed by BSTFA (79.2
mL, 0.30
mmol, 1.5 equiv), were added to a solution of 2b (Rl=Me, R2=Me) prepared by
Method C (50
mg, 0.20 mmol, 1 equiv) in dry DMF (0.5 mL) at 0°C. The reaction
mixture was stirred at 0°C
for 10 min, and then at rt for 50 min. The reaction mixture was added to 8c
(R9=3-(4-
chlorophenyl)propyl) prepared by Method M (97.3 mg, 0.26 mmol, 1.3 equiv) in a
25 mL round
bottom flask, followed by HATU (123 mg, 0.32 mmol, 1.6 equiv). The reaction
mixture was
stirred at rt for 3 h. The reaction mixture was evaporated to dryness, taken
up in ethyl acetate
(60 mL), and washed with 10% citric acid (2 x 40 mL), water (40 mL), half sat.
NaHCO3 (40
mL), and brine. The organic layer was dried over Na2S04 and evaporated to give
a yellow
syrup.
[0265] Trifluoroacetic acid (3 mL) and water (0.2 mL) were added to a solution
of the above
syrup in dichloromethane (9 mL) with methyl sulfide (0.2 mL). The reaction
mixture was
stirred at rt for 1 h. The solvent was removed under vacuum and co-evaporated
with toluene
twice. The residue was purified by chromatography to provide 1-(2-(S)-4-(R)-(3
p-
chlorophenyl)prop-1-ylpyrrolidin-2-yl)-N f 1-(S)-[2-(S)-3-(S), 4-(S), 5-(R)-
trihydroxy-6-(R)-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (41.6 mg, 42%)
as a white
solid.
[0266] 1H NMR (300 MHz, CDC13) S 7.26-7.21 (m, 2), 7.17-7.12 (m, 2), 5.23 (d,
J= 5.7, 1),
4.10-4.00 (m, 3), 3.83-3.75 (m, 1), 3.74-3.70 (m, 1), 3.54-3.48 (m, 1), 3.25-
3.18 (m, 1), 2.63-
2.50 (m, 3), 2.20-2.00 (m, 3), 2.09 (s, 3), 1.85-1.74 (m, 1),1.68-1.55 (m, 2),
1.42-1.33 (m, 2),
0.95-0.85 (m, 6). MS(ESPOS): 501.5 [M + H]+ MS(ESNEG): 499.4 [M - H]-.
74



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 16
Preparation of 1-[2-(S)-4-(S)-(2,2-difluoropent-1-yl)pyrrolidin-2-yl]-N {1-
[3,4,5-
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl]acetamide
<'~ O
F F
H~,.
H ~~6Me
HO ~H
[0267] Triethylamine (88.3 mL, 0.64 moral, 3.2 equiv), followed by BSTFA (79.2
mL, 0.30
mmol, 1.5 equiv), were added to a solution of 2b (Rl=Me, R2=Me) prepared by
Method C (50
mg, 0.20 mmol, 1 equiv) in dry DMF (0.5 mL) at 0°C. The reaction
mixture was stirred at 0°C
for 10 min, and then at rt for 50 min. The reaction mixture was added to the
acid 9d (R9=2,2-
difluoropentyl) prepared by Method O (67.7 mg, 0.21 mmol, 1.1 equiv) in a 25
mL round
bottom flask, followed by HATU (101 mg, 0.27 mmol, 1.3 equiv). The reaction
mixture was
stirred at rt for 3h. The reaction mixture was evaporated to dryness, taken up
in ethyl acetate (60
mL), and washed with 10% citric acid (2 x 40 mL), water (40 mL), half sat.
NaHC03 (40 mL),
and brine. The organic layer was dried over Na2S04 and evaporated to give a
yellow syrup.
[0268] Trifluoroacetic acid (3 mL) and water (0.20 mL) were added to a
solution of the
above syrup in dichloromethane (9 mL) with methyl sulfide (0.20 mL). The
reaction mixture
was stirred at rt for 1 h. The solvent was removed under vacuum and co-
evaporated with
toluene twice. The residue was purified by chromatography to provide 1-[2-(S)-
4-(S)-(2,2-
difluoropent-1-yl)pyrrolidin-2-yl]-N { 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-
2-methylprop-1-yl}acetamide (56 mg, 62%) as a white solid.
[0269] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J= 5.7, 1), 4.17-4.04 (m, 3), 3.98
(dd, J=
3.3, 9.3, 1), 3.77 (d, J= 3, 1), 3.51 (dd, J= 3.4, 10.3, 1), 3.40 (dd, J= 6.9,
10.5, 1), 2.71 (dd, J=
10.2, 10.2, 1), 2.42-2.33 (m, 1), 2.23-2.11 (m, 2), 2.10 (s, 3), 2.08-1.73 (m,
5), 1.56-1.42 (m, 2),
0.99-0.89 (m, 9). MS(ESPOS): 455.5 [M + H]+; MS(ESNEG): 453.5 [M - H]-.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 17
Preparation of 1-(-4-n-propylpiperid-6-yl)-N ~1- [3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
SMe
[0270] Triethylamine (0.18 mL, 1.26 mmol) and BSTFA (0.549 mL, 2.1 mmol) were
added
to the lincosamine intermediate 2b (Rl=Me, Ra=Me) prepared by Method D (102
mg, 0.42
mmol) in DMF (5 mL) at 0°C, and the mixture was stirred at rt for 3 h.
Acid 10b (R~=propyl)
prepared by Method P (200 mg, 0.84 mmol) and HATU (319 mg, 0.84 mmol) were
added and
the mixture was stirred for 4 h at rt. DMF was removed and the residue was
extracted with ethyl
acetate (100 mL) and washed with saturated bicarbonate (40 mL). The product
obtained by
removal of solvent was taken up in methanol and treated with Dowex H+ resin
for 1 h. After
filtering the resin, methanol was removed to obtain the crude product. The
crude product was
then purified on silica gel column using 10% methanol in dichloromethane as
the eluent to
provide pyridine llb (Rl=Me, R2=Me, R3=H, R9~ropyl) (117 mg, 58%).
[0271] TLC: Rf= 0.81 (10% methanol in dichloromethane); 1H NMR (300 MHz,
CDCl3) b
1.20 (t, J= 6.3, 6H), 2.19 (m, 2), 2.32 (s, 3), 2.43 (m, 1}; 2.84-2.97 (m, 4),
3.74 (m, 1), 4.06 (m,
1), 4.31 (m, 1), 4.52 (m, 2), 5.42 (d, J= 5.7, 1), 7.33-7.61 (m, 5), 7.80 (m,
1), 8.15 (s, 1), 8.69 (d,
J= 4.8, 1); MS (ESPOS): 475 (M+H).
[0272] Pt02 (100 mg, 0.44 mmol) was added to pyridine llb (Rl=Me, R2=Me, R3=H,
R9=propyl), (150 mg, 0.37 mmol) in methanol (2 mL), water (lOmL), and acetic
acid (3 mL),
and the mixture was hydrogenated at 50 psi overnight. The product obtained
after filtering the
catalyst and removing the solvent was purified by silica gel column
chromatography using 30%
methanol in dichloromethane as the eluent to provide the title compound (20
mg, 14%).
[0273] TLC: Rf= 0.7 (50% methanol in dichloromethane);1H NMR (300 MHz, CD30D)
8
5.24 (d, J= 6.9, 1), 4.86 (m, 1), 4.13 (m, 2), 3.79 (d, J= 3.3, 1), 3.52 (dd,
J= 3.3, 9.9, 1), 3.32
(m, 1), 3.17 (m, 1), 2.67 (m, 1), 2.17 (m, 1), 2.10 (s, 3), 1.97 (m, 1), 1.74
(m, 1), 1.54 (m, 1),
1.38 (m, 2), 1.31 (m, 2), 1.14 (m, 2), 1.02 (m, 9) MS(ESPOS): 405 (M+H).
76



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 18
Preparation of 1-(2-(S)-4-(R)-n-pentyl-N (2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N
~1-[3,4,5
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
'' O
Hf~
H ~~~6Me
OH .
HO ~H
[0274] DIEA (0.1 mL, 0.57 mmol) and liquid ethylene oxide (3 mL) were added to
a stirred
solution of crude 1-(-4-h-pentylpyrrolidin-2-yl)-N f 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide, prepared as in
Example 10
(237.4 mg), in anhydrous methanol (10 mL), at 0°C and under nitrogen.
The resulting solution
was stirred at -4°C for 18 h and evaporated to dryness. The residue
obtained was purified by
chromatography over silica gel with an eluent of 5% methanol ammonia in
dichloromethane.
The desired fractions were evaporated and lyophilized (deuterium
oxide/anhydrous acetonitrile,
l:l, v/v, lOmL) to furnish the title compound as a fluffy white powder
(SO.lmg, 30.2%); TLC,
Rf= 0.68 (14% methanolic ammonia in dichloromethane);1H NMR (300 MHz) S 5.40
(d, J=5.8,
1), 4.55 (m, 1), 4.24 (s, 1), 4.17-4.11 (m, 1), 3.99-3.89 (m, 4), 3.69-3.65
(m, 1), 3.47 (d, J=4.4,
2), 3.01 (m, 1), 2.33 (br s,4), 2.18 (s,4), 1.57-1.32 (m,9), 0.94-0.87 (m, 9).
MS(ESPOS):464[M+H];(ESNEG):497.5[M-H+HCl].
Example 19
Preparation of 1-[2-(S)-4-(R)-n-pentyl-N (2-(R)-methyl-2-hydroxyeth-1-
yl)pyrrolidin-2-yl]-
N f 1-(3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide
77



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
O
HN,~.
H ~~6Me
OH ._
[0275] DIEA (O.lmL, 0.58 mmol, 1 equiv) and R(+)-propylene oxide (3 mL) were
added to
a stirred cool solution of crude 1-(2-(S)-4-(R)-h-pentylpyrrolidin-2-yl)-N ~ 1-
[3,4,5-trihydroxy-
6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (307.6 mg,
0.58 mmol, 1
equiv), prepared as in Example 10, in anhydrous methanol (lOmL), at 0°C
and under nitrogen.
The resulting solution was stirred at -4°C for 18 h and evaporated to
dryness. The residue
obtained was purified by chromatography over silica gel, with an eluent of 6%
methanol
ammonia in dichloromethane. The desired fractions were evaporated, and
lyophilized
(deuterium oxide/anhydrous acetonitrile, 1:1, v/v, 20mL) to furnish the title
compound as a
fluffy white powder (9lmg, 48%
[0276] TLC, Rf= 0.7 (14% methanolic ammonia in dichloromethane); 1H NMR(300
MHz,
CD30D) 8 5.44 (d, J= 5.5, 1), 4.31 (s, 2), 4.26-4.11 (m, 1), 3.97 (d, J=
3.3,1.1,1), 3.75 (dd, J=
3.3,3.3, 1), 3.39 (dd, J= 3.8,3.8, 1), 2.31 (s, 3), 1.5-0.95 (m, 12), 1.34 (d,
J= 6.0, 4), 1.17-1.10
(m, 13); MS(ESPOS): 477.6 [M+H], (ESNEG): 475.6 [M-H].
Example 20
Preparation of 1-[2-(S)-4-(R)-n-pentyl-N (2-(S)-methyl-2-hydroxyeth-1-
yl)pyrrolidin-2-yl]-
N f 1-[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl]acetamide
/' O
H1~
H ~~~6Me
OH
HO ~H
[0277] Dimethyl sulfide (62 p.L), TFA (1 mL), and water (62 ~,L) were added to
a stirred
solution of the Boc-protected 1-(2-(S)-4-(R)-n-pentylpyrrolidin-2-yl)-N f 1-
[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl]acetamide (92mg, 0.18
mmol), prepared
as in Example 10, in anhydrous dichloroethane (3 mL). The resulting solution
was stirred at rt
78



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
for 1 h and evaporated to dryness. Anhydrous methanol (8 mL) and DIEA (31 ~,L,
0.18 mmol)
were added to the residue obtained. The mixture was cooled to -4°C and
S-(-)-propylene oxide
(2 mL) was added. The resulting solution was stirred at -4°C for 18 h,
evaporated to dryness,
and purified by chromatography over silica gel, with an eluent of 6% methanol
ammonia in
dichloromethane. The desired fractions were evaporated and lyophilized
(deuterium/anhydrous
acetonitrile, 1:1, vlv, 8 mL) to furnish the title compound as a fluffy white
powder (29.8 mg,
31.2%).
[0278] TLC, Rf= 0.7 (12% methanolic ammonia in dichloromethane); 1H NMR (300
MHz,
CD30D) 8 5.44 (d, J= 5.5,1), 4.35-4.19 (m, 4), 4.02 (d, J= 3.3, 2), 2.75 (d,
J= 6.3,2.2, 3), 2.3
(s, 3), 1.50 (m, 11), 1.4 (d, J= 6.0, 3.5, 3), 1.16-1.10 (m, 12). MS(ESPOS):
477.6 [M+H];
(ESNEG) 475.4 [M-H].
Example 21
Preparation of 1-(-4-n-pentyl-N(3-hydroxyprop-1-yl)pyrrolidin-2-yl)-N~1-[3,4,5-

trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl]acetamide
HH,.
O
H ~~6Me
HO ~ HO 'OH
[0279] Triethylamine (0.2 mL, 1.38 mmol, 3 equiv), followed by 3-bromo-1-
propanol (60
~,L, 0.69 mmol, 1.5 equiv), were added to a stirred solution of crude 1-(2-(S)-
4-(R)-h-
pentylpyrrolidin-2-yl)-N {1-(R)- [2-(S)-3-(S), 4-(S), 5-(R)-trihydroxy-6-(R)-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (192.5 mg, 0.46
mmol, 1
equiv), prepared as in Example 10, in anhydrous acetonitrile (2 mL), under
nitrogen. The
resulting mixture was stirred at rt for 18 h and evaporated to dryness. The
residue obtained was
purified by chromatography over silica gel with an eluent of 5% methanol
ammonia in
dichloromethane. The desired fractions were pooled together, evaporated to
dryness, and
lyophilized to furnish the title compound as a white fluffy powder (13.5 mg,
6%).
[0280] TLC, Rf= 0.75 (14% methanolic ammonia in dichloromethane);1H NMR (300
MHz,
CD30D) 8 5.44 (d, J= 5.8, 1), 4.33-4.26 (m, 4), 4.01 (d, J= 2.7, 1), 3.85-3.74
(m, 6), 2.29 (s, 3),
2.1 (m, 4), 1.54 (m, 8), 1.16-1.08 (m, 12); MS (ESPOS): 477.6 [M+H].
79



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 22
Preparation of 1-[2-(S)-4-(R)-(3-methylbut-1-yl)-N (2-hydroxyeth-1-
yl)pyrrolidin-2-yl]-N
{1-[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide
~\ ~O
Hf~
H ~~6Me
OH
HO ~~ H
[0281] Ethylene oxide (0.6 mL) was added to a solution of 1-[2-(S)-4-(R)-(3-
methylbut-1-
yl)pyrrolidin-2-yl]-N { 1-(R)- [2-(S)-3-(S), 4-(S), 5-(R)-trihydroxy-6-(R)-
(methylthio)-
tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide (35.1 mg, 0.084 mmol),
prepared as in
Example 9, in methanol (3 mL), at 0°C. The reaction mixture was stirred
at 4°C overnight.
Additional ethylene oxide (0.6 mL) was added and stirred at 4°C
overnight. The reaction
mixture was concentrated and purified by chromatography to give a white solid,
1-[2-(S)-4-(R)-
(3-methylbut-1-yl)-N (2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N { 1-(R)- [2-(S)-3-
(S), 4-(S), 5-(R)-
trihydroxy-6-(R)-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
(24.1 mg,
62%).
[0282] 1H NMR (300 MHz, CDC13) 8 7.68 (d, J= 9.0, 1), 5.32 (d, J= 5.4, 1),
5.24 (d, J=
3.0, 1), 4.13-4.07 (m, 1), 4.01 (ddd, J= 2.8, 9.9, 9.9, 1), 3.86 (d, J=10.8,
1), 3.78-3.68 (m, 2),
3.61-3.57 (m, 1), 3.56-3.48 (m, 1), 3.36-3.32 (m, 1), 3.27-3.21 (m, 1), 2.94-
2.85 (m, 1), 2.76-
2.70 (m, 1), 2.55 (ddd, J= 3.6, 3.6, 12.6, 1), 2.41-2.37 (m, 1), 2.36-2.27 (m,
1), 2.15 (s, 3), 2.03-
1.95 (m, 2), 1.93-1.81 (m, 1), 1.54-1.42 (m, 1), 1.39-1.26 (m, 2), 1.22-1.10
(m, 2), 0.99-0.92 (m,
6), 0.90-0.84 (m, 6). MS(ESPOS): 463.5 [M + H] + MS(ESNEG): 461.5 [M - H]-.



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 23
Preparation of 1-[4- (3,3-difluoroprop-1-yl)-N (2-hydroxyeth-1-yl)pyrrolidin-2-
yl]-N {1
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide
~~O
('N~H~,.
H ~~6Me
OH
HO 'O H
[0283] Ethylene oxide (0.4 mL) was added to a solution of 1-[2-(S)-4-(R)-(3,3-
difluoroprop-
1-yl)pyrrolidin-2-yl]-N { 1-(R)- [2-(S)-3-(S), 4-(S), 5-(R)-trihydroxy-6-(R)-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl]acetamide, prepared as in
Example 14
(29.7 mg, 0.07 mmol), in methanol (2 mL), at 0°C. The reaction mixture
was stirred at 4°C
overnight. Additional ethylene oxide (0.4 mL) was added and stirred at
4°C overnight. The
reaction mixture was concentrated and purified by chromatography to give a
white solid, 1-[2-
(S)-4-(R)-(3-methylbut-1-yl)-N (2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N {1-(R)-
[2-(S)-3-(S), 4-
(S), 5-(R)-trihydroxy-6-(R)-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl}acetamide
(19.3 mg, 59%).
[0284] 1H NMR (300 MHz, CDC13) ~ 5.86 (dddd, J= 4.3, 4.3, 57, 57, 1), 5.23 (d,
J= 5.7, 1),
4.13-4.04 (m, 3), 3.75 (d, J= 3.3, 1), 3.73-3.57 (m, 2), 3.53 (dd, J= 3.3,
10.2, 1), 3.42-3.36 (m,
1), 3.26-3.18 (m, 1), 2.88-2.78 (m, 1), 2.62-2.55 (m, 1), 2.17-2.00 (m, 4),
2.10 (s, 3), 1.94-1.73
(m, 3), 1.55-1.45 (m, 2), 0.98-0.91 (m, 6).
MS (ESPOS): 471.5 [M + H]+, MS (ESNEG): 469.4 [M - H]-.
81



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 24
Preparation of 1-[2-(S)-4-(R)-m-pentyl-N (2-hydroxyeth-1-yl)pyrrolidin-2-yl]-N
{1-[3,4,5
trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2,2-difluoroprop-1-
yl~acetamide
" s° O F
F
Hf~
H -~i6Me
OH
HO ~H
[0285] Ethylene oxide (1 mL, excess) was added to the title compound from
Example 12 (60
mg, 0.136 mmol) in methanol (5 mL) at 0°C, and the mixture was stirred
at 4°C overnight. The
solvent was removed and the crude product was purified by silica gel column
chromatography
using 10% methanol in dichloromethane as the eluent (25 mg, 38%).
[0286] TLC: Rf= 0.76 (5% methanol in dichloromethane); 1H NMR (300 MHz, CD3OD)
8
0.89 (m, 3), 1.30 (m, 8), 1.68 (t, J=19.4, 3), 1.78 (m, 1), 1.99 (m, 2), 2.07
(s, 3), 2.63 (m, 1),
2.73 (m, 1), 3.19 (m, 1), 3.58 (m, 3), 3.95 (m, 1), 4.08 (dd, J= 6.0, 9.90,
1), 4.44-4.60 (m, 2),
5.26 (d, J= 5.4, 1); MS (ESPOS): 485 (M+H).
Example 25
Preparation of 1-(4-n-pentylpiperid-6-yl)-N ~ [3, 4, 5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl) acetamide
O
NH HI~~
H ~~6Me
HOr~H
[0287] 4-Pentylpyridine-2-carboxylic acid (lOb) (R9=pentyl) was made by
employing
general Method P. To 4-pentylpyridine (3 g, 20 mmol) in acetic acid (30 mL),
hydrogen
peroxide (0.7 g, 30%, 20 mmol) was added and refluxed overnight. Removal of
solvent resulted
in residue which was dissolved in DCM (100 mL) dried over MgS04 and filtered.
Removal of
82



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
DCM resulted in a brown liquid, 4-pentylpyridine-N oxide, (3.3 g, 100%). To
trimethylsilyl
cyanide (2.37 g, 24 mmol), 4-pentylpyridine N oxide (3.3 g, 20 mmol) in DCM
(10 mL) was
added followed by dropwise addition of dimethylcarbamoyl chloride (2.56 g, 24
mmol) in DCM
(10 mL). After stirring at room temperature overnight, sodium bicarbonate (100
mL,10 %) was
added and the organic layer was separated. The aqueous layer was extracted
twice with DCM
(50 mL) and the combined organic layer was dried over magnesium sulfate.
Removal of solvent
resulted in compound l0a (R9~entyl) (4.1 g, 100%).
[0288] 1H NMR (300 MHz, CD30D) 8 8.52 (m, 1), 7.46 (s, 1), T.27 (m, 1), 3.00
(m, 2), 2.60
(m, 2), 1.60 (m, 2), 1.27 (m, 2), 0.86 (m, 3). MS (ES+): 175 (M+1).
[0289] The 4-Pentyl-2-cyanopyridine (l0a) (R9=pentyl) (3.4 g, 19.5 mmol) from
the
previous step was dissolved in HCl (6 N, 100 mL) and refluxed overnight. The
residue obtained
on removal of HCl was purified by column chromatography using 20% MeOH in DCM
(3.7 g,
100%) to give product compound 10(b). 1H NMR (300 MHz, CD30D) 8 8.74 (d, J--
6.3, 1), 8.39
(s, 1), 8.06 (d, J 6.3, 1), 2.98 (t, J--7.2, 2), 1.77(m, 2), 1.39 (m, 4), 0.95
(t, J--7.2, 3H). MS (ES-
): 192 (M-1).
[0290] Then to 7-methyl oc-thiolincosaminide 2b (Rl=Me, R2=Me) (90 mg, 0.35
mmol) in
DMF (2ml), TEA (72 mg, 0.7 xnmol), BSTFA (276 mg, 1.05 mmol) were added at 0
°C and
stirred at room temperature for 1.5 hr. Then the acid (lOb) (R9~entyl) (138
mg, 0.7 mmol) and
HATU (165 mg, 0.53 mmol) was added to the reaction mixture, and stirred at
room temperature
overnight. DMF was completely removed, the residue was taken up in EtOAc (50
mL), washed
with sodium bicarbonate (10%, 50 mL), brine (50 mL). The product obtained
after drying over
magnesium sulfate and concentration was taken up in methanol (10 mL) and
treated with NR-50
resin (150 mg) for 3 hr. The resin was filtered and the solvent was removed.
Purification of the
crude product was carried out silica gel column chromatography using 3% MeOH
in DCM as
eluent to obtain compound llb (Rl=Me, R2=Me R3=H, R9=pentyl) (90 mg, 59%):
[0291] 1H NMR (300 MHz, CD30D) 8 8.50 (d, J--5.1, 1), 7.95 (s, 1), 7.11 (m,
1), 5.25 (d,
J--5.7, 1), 4.20-4.87 (m, 3), 3.85 (d, J 3.3, 1), 3.55 (dd, J--3.3, 7.2, 1),
2.72 (m, 2), 2.16 (m, 4),
1.67 (m, 2), 1.35 (m, 4), 0.96 (m, 9). MS (ES+): 427 (M+1).
[0292] To pyridine llb (Rl=Me, R2=Me R3=H, R9~entyl) (90 mg, 0.7 mmol) in
water (10
mL), AcOH (3 mL) and MeOH (2 mL), Pt02 (100 mg) was added, hydrogenated at 55
psi
overnight. The solvent was removed to obtain the crude product. Purification
of the crude
product was carried out by silica gel column chromatography using 20% MeOH in
DCM to
obtain the title compound (35 mg, 38%).
83



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0293] 1H NMR (300 MHz, CD30D) 8 5.23 (d, J--5.1, 1), 4.17 (m, 3), 3.79 (d, J--
3.3, 1),
3.52 (m, 1), 3.38 (m, 1), 3.07 (m, 1), 2.68 (m, 1), 2.14 (m, 4), 1.88 (m, 1),
1.71 (m, 1), 1.52 (m,
1), 1.30 (m, 8), 1.07 (m, 3), 0.90 (m, 9); MS (ES+): 433 (M+1).
Example 26
Preparation of 1-(4-methoxypiperid-6-yl)-N ( [3, 4, 5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl)acetamide
O
NH Hf~
HC~~ ~ ~SMe
HOY~'OH
[0294] 4-Methoxypyridine-2-carboxylic acid, compound 10(b) (R9 = methoxy), was
made
employing general Method P. To trimethylsilyl cyanide (0.95 g, 9.6 mmol), 4-
methoxypyridine
N oxide (1 g, 8 mmol) in DCM (10 mL) was added, followed by dimethylcarbamoyl
chloride
(1.03 g, 9.6 mmol) in DCM (10 mL), dropwise. After stirring at room
temperature overnight,
sodium bicarbonate (100 mL, 10%) was added, and the organic layer was
separated. The
aqueous layer was extracted twice by DCM (50 mL each). The combined organic
layer was
dried over magnesium sulfate and the solvent was removed to obtain product,
compound 10a
(0.97 g, 90%):
[0295] 1H NMR (300 MHz, CD30D) 8 8.52 (m, 1), 7.22 (m, 1), 7.01 (m, 1), 3.92
(s, 3H);
MS (ES+): 135 (M+1).
[0296] 4-Methoxy-2-cyanopyridine, compound l0a (R9 = methoxy), (0.97 g, 7.2
mmol) was
dissolved in HCl (6N, 50 mL), and refluxed overnight. The HCl was evaporated
and the
resulting product was crystallized from acetonile, to give compound lOb
(R9=methoxy) (0.6 g,
60%).
[0297] iH NMR (300 MHz, CD3OD) 8 8.65 (m, 1), 7.99 (m, 1), 7.68 (m, 1), 4.21
(s, 3H).
MS (ES-): 152 (M-1).
84



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0298] To 7-methyl a-thiolincosaminide, compound 2b (Rl=Me, R2=Me), (90 mg,
0.35
mmol) in DMF (2 mL), TEA (72 mg, 0.7 mmol), BSTFA (276 mg, 1.05 mmol) were
added at
0°C and left stirred at room temperature for 1.5 hr. Then compound lOb
(R9 = methoxy) (109
mg, 0.7 mmol) and HATU (165 mg, 0.53 mmol) were added to the reaction mixture,
and stirred
at room temperature overnight. The DMF was completely removed and the residue
was taken up
in EtOAc (50 mL), washed with sodium bicarbonate (10%, 30 mL); brine (30 mL),
and dried
over magnesium sulfate. The solvent was removed to obtain a brown oil-like
liquid, which was
dissolved in methanol (10 mL) and treated with NR-50 resin for 1 hr. The resin
was filtered, and
the solvent was removed to obtain the crude material. Purification was carried
out on silica gel
column chromatography using EtOAc as eluent to obtain compound 11b (Rl=Me,
Ra=Me R3=H,
R9=methoxy) (100 mg, 72%).
(0299] 1H NMR (300 MHz, CD30D) 8 8.42 (m, 1 ), 7.64 (m, 1 ), 7.07 (m, 1 ),
5.25 (d, J--5.4,
1), 4.07-4.87 (m, 3), 3.94 (m, 4), 3.56 (m, 1), 2.99 (m, 2), 2.80 (m, 1), 2.22
(m, 1), 2.11 (s, 3),
0.96 (m, 3). MS (ES+): 387 (M+1).
[0300] To compound 11b (Rl=Me, R2=Me R3=H, R9=methoxy) (100 mg, 0.26 mmol) in
water (10 mL), AcOH (3 mL) and MeOH (2 mL), PtO2 (100 mg) were added and
hydrogenated
at 55 psi overnight. The solvent was removed to obtain the crude product.
Purification of the
crude product was carried out by silica gel column chromatography using 20 %
MeOH in DCM
to obtain the title compound. (9 mg, 9%).
[0301] 1H NMR (300 MHz, CD30D) b 5.24 (d, J--5.7, 1), 4.17 (dd, J--9.9; 3.3,
1), 4.07 (m,
2), 3.79 (m, 1), 3.52 (dd, J--10.5; 3.3, 1), 3.35 (s, 3), 3.18 (m, 2), 2.72
(m, 1), 2.16 (m, 1), 2.12
(s, 3) 1.99 (m, 2), 1.50 (m, 1), 1.24 (m, 2), 0.90 (d, J--6.9, 6); MS (ES+):
393 (M+1).
Example 27
Preparation of 1-[4-(1-ethylprop-1-yl)piperid-6-yl]-N ~1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
~~~SMe
)H
[0302] 4-Isopentylpyridine-2-carboxylic acid, compound lOb (R9 =1-ethyl-
propyl), was
made by employing general Method P. To 4-(1-ethyl-propyl)-pyridine (8.5 g, 57
mmol) in
acetic acid (30 mL), hydrogen peroxide (17.8 g, 30 %, 57 mmol) was added. The
resulting
reaction mixture was refluxed overnight. The residue obtained on removal of
solvent was
dissolved in DCM (100 mL), dried over MgSO4. After filtering, the solvent was
removed to
obtain a brown liquid, 4-(1-ethyl-propyl)-pyridine-N oxide (9 g, 95%).
[0303] To a solution of trimethylsilyl cyanide (6.5 g, 65 mmol) and 4-(1-ethyl-
propyl)-
pyridine-N oxide (9 g, 54 xnmol) in DCM (25 mL) was added a solution of
dimethylcarbamoyl
chloride (7 g, 65 mmol) in DCM (10 mL), dropwise. After stirring at room
temperature
overnight, sodium bicarbonate (100 mL, 10%) was added, and the organic layer
was separated.
The aqueous layer was extracted twice with DCM (50 mL). The combined organic
layer was
dried over magnesium sulfate and the solvent was removed to obtain the
product, compound l0a
(R~ =1-ethyl-propyl) (9.6 g, 100%).
[0304] 1H NMR (300 MHz, CD30D) ~ 8.58 (m, 1), 7.46 (m, 1), 7.26 (m, 1), 2.42
(m, 1),
1.77 (m, 4)), 0.78 (t, J--7.5, 6). MS (ES+): 175(M+1).
[0305] Compound l0a (R9 =1-ethyl-propyl) (9.5 g, 54 mmol) was dissolved in HCl
(6N, 50
mL) and refluxed overnight. HCl was evaporated and the resulting product,
compound lOb (R9
= 1-ethyl-propyl), was crystallized from acetonitrile (10 g; 100%).
[0306] 1H NMR (300 MHz, CD3OD) 8 8.86 (m, 1), 8.45 (m, 1), 8.20 (m, 1), 2.92
(m, 1),
1.87 (m, 4), 0.84 (t, J--7.5, 6). MS (ES-): 192 (M-1).
[0307] To the acid lOb (77 mg, 0.4 mmol) in DMF (2 mL), 7-methyl a-
thiolincosaminide,
compound 2b (Rl=Me, Ra=Me), (100 mg, 0.4 mmol) was added, followed by HBTU
(166 mg,
0.44 mmol) and DIEA (205 mg, 0.8 mmol). The mixture was stirred at room
temperature for 2
hr. The product was obtained on removal of DMF and purified by silica gel
column
chromatography using ethyl acetate to provide compound llb (Rl=Me, R2=Me R3=H,
R9= 1-
ethyl-propyl) (150 mg, 89%).
86



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0308] 1H NMR (300 MHz, CD30D) ~ 8.42 (d, J--5.1, 1), 7.37 (s, 1), 7.32 (m,
1), 5.27 (d,
J--4.8,1 ), 4.21-4.8 8 (m, 3), 3 .85 (d, J--3.6, 1 ), 3 .5 6 (dd, J--3 .3,
10.2, 1 ), 2.48 (m, l ), 2.11 (m, 4),
1.00 (m, 12). MS (ES+): 427 (M+1).
[0309] To compound llb (Rl=Me, R2=Me R3=H, R9= 1-ethyl-propyl) (130 mg, 0.3
mmol)
in water (10 mL), AcOH (2 mL) and MeOH (2 mL), Pt02 (150 mg) was added and
hydrogenated at 55 psi overnight. The solvent was removed to obtain the crude
product.
Purification was carried by silica gel column chromatography using 20% MeOH in
DCM to
obtain the title compound (40 mg, 30%).
[0310] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J--5.7, 1), 4.17 (dd, J--9.9; 3.3,
1), 4.10 (m,
2), 3.78 (m, 1), 3.51 (m, 2), 2.81 (m, 2), 2.16 (m, 1), 2.10 (s, 3) 1.90 (m,
2), 1.76 (m, 3), 1.40 (m,
8), 0.91 (m, 9); MS (ES+): 433 (M+1).
Example 28
Preparation of 1-(4-iso-propylpiperid-6-yl)-N ~ 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl)acetamide
NH HNi
H ~ ~ 6Me
HO ~H
[0311] 4-Isopropylpyridine-2-carboxylic acid, compound (lOb) (R9 = isopropyl),
was made
by employing general Method P. To 4-isopropylpyridine (5 g, 0.041 mol) in
acetic acid (60 mL),
hydrogen peroxide (30%, 4.7 g, 0.13 mol) was added and refluxed over night.
After removing
the solvent, the residue was dissolved in DCM dried over magnesium sulfate and
taken as such
for the next step. To the resulting compound in dichloromethane (10 mL)
trimethylsilyl cyanide
(7.0 mL, 0.07 mol) and dimethylcarbamyl chloride (5.6 mL, 0.05 mol) were added
and stirred at
room temperature for 24 hours. Aqueous potassium carbonate (10%, 50 mL) was
added and
extracted with dichloromethane (100 mL). The crude product obtained on removal
of solvent
was taken up in hydrochloric acid (6N, 30 mL) and refluxed for 24 hours.
Removal of acid
87



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
followed by crystallization of the crude product from acetonitrile resulted in
acid lOb (R9 =
isopropyl) (Sg, 75%).
[0312] 1H NMR (300 MHz, CD30D): 8 8.78 (d, J = 6, 1), 8.42 (s, 1), 8.16 (d, J=
6.0, 1),
3.25 (m, 1), 1.33 (d, J= 9.0, 6) MS (ES-): 164 (M-1).
[0313] To the amine, compound 2b (Rl=Me, R2=Me), (140 mg, 0.56 moral) in DMF
(3 mL),
BSTFA (0.59 mL, 2.24 mmol) and triethylamine (0.18 mL, 1.26 mmol) were added
at 0°C and
the reaction mixture was stirred at room temperature for 3 hours. Acid lOb (R9
= isopropyl)
(188 mg, 1.13 mmol) and HATU (319 mg, 0.84 mmol) were combined and left
stirred for
further 4 hours at room temperature. The DMF was removed and the residue was
extracted with
ethyl acetate (100 mL) and washed with saturated bicarbonate (40 mL). The
product obtained on
removal of solvent was taken up in methanol and treated with Dowex H+ resin
for 1 hour. After
filtering the resin, methanol was removed to obtain the crude product. It was
then purified on
silica gel column using 10% methanol in dichloromethane as eluent to provide
compound llb
(Rl=Me, R~=Me R3=H, R~ = isopropyl) (120 mg, 53 %).
[0314] 1H NMR (300 MHz, CD30D): 8 8.42 (d, J--5.1, 1), 7.37 (s, 1), 7.32 (m,
1), 5.27 (d,
J 4.8,1), 4.21-4.88 (m, 3), 3.85 (d, J--3.6, 1), 3.56 (dd, J 3.3, 10.2, 1),
2.48 (m, 1), 2.11 (m, 1),
2.10 (s, 3), 1.20 (m, 12). MS (ES+): 399 (M+1).
[0315] To llb (Rl=Me, R2=Me R3=H, R9 = isopropyl) (100 mg, 0. 257 mmol) in
methanol
(5 mL), water (10 mL) and acetic acid (5 mL), platinum dioxide (100 mg, 0.44
mmol) was
added and hydrogenated at 60 psi for 16 h. After filtering the catalyst, the
solvent was stripped
off to obtain the crude product which was then purified on silica gel column
chromatography
using 10% methanol in dichloromethane as eluent. The lower Rf compound was the
title
compound (10 mg, 9%).
[0316] 1H NMR (300 MHz, CD30D): 8 5.24 (d, J--5.7, 1), 4.17 (dd, J=9.9; 3.3,
1), 4.10 (m,
2), 3.80 (m, 1), 3.51 (m, 1), 3.16 (m, 1), 2.61 (m, 1), 2.16 (m, 1), 2.10 (s,
3) 1.90 (m, 1), 1.76 (m,
1),1.50-1.09 (m, 5), 0.91 (m, 12); MS (ES+): 405 (M+1).
Example 29
Preparation of 1-(4-n-butylpiperid-6-yl)-N f 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
88



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
NH HN ~~
H ~ ~ ~SMe
HOr~H
[0317) 4-Butylpyridine was prepared by adding potassium-t-butoxide (0.68 g, 6
mmol) to
propylphosphonium bromide (Aldrich) (2.4 g, 6.0 mmol) in THF (10 mL), at 0
°C and stirring at
room temperature for 1 hour. Pyridine-4-caxbaldehyde (428 mg, 4 mmol) was
added and the
reaction mixture stirred for 2h. The reaction mixture was then poured into
water and extracted
with ethyl acetate. The product obtained after removing the solvent was taken
as such in
methanol (30 mL) to which palladium on carbon (10%, 300 mg) was added and
hydrogenated at
1 atm pressure over night. Removal of solvent and purification on column
chromatography
using ethyl acetate resulted in pure product 4-butylpyridine (500 mg, 92%):
[0318] 1H NMR (CDC13): S 8.42 (d, J= 6.0, 2), 7.05 (d, J= 6.0, 1), 2.60 (t, J=
6.5, 2), 1.62
(m, 2), 1.37 (m, 2), 0.93 (t, J= 7.0, 3). MS (ES+): 136 (M+1).
[0319] 4-Butylpyridine-2-carboxylic acid, compound (lOb) (R9 = butyl), was
made
employing general Method P. To 4-butylpyridine (2 g, 0.014 mol) in acetic acid
(15 mL),
hydrogen peroxide (30%, 5 mL, 0.056 mol) was added and refluxed over night.
After removing
the solvent, the residue was dissolved in DCM dried over magnesium sulfate and
taken as such
for the next step. To the compound from the previous step in dichloromethane
(10 mL)
trimethylsilyl cyanide (3.92 mL, 0.029 mol) and dimethylcarbamoyl chloride
(2.67 mL, 0.028
mol) was added and stirred at room temperature for 24 hours. Aqueous potassium
carbonate
(10%, 50 mL) was added and extracted with dichloromethane (100 mL). The crude
product
obtained on removal of solvent was taken up in hydrochloric acid (6N, 30 mL)
and refluxed for
24 hours. Removal of acid followed by crystallization of the crude product
from acetonitrile
resulted in acid lOb (R9 = butyl) (l.Sg, 60%).
[0320] iH NMR (CDCl3): 8 8.92 (d, J= 6.0, 1), 8.65 (s, 1), 8.27 (m, 1), 3.23
(t, J= 6.5, 2),
1.98 (m, 2), 1.67 (m, 2), 1.20 (t, J= 7.0, 3). MS (ES-): 178 (M-1).
[0321] To the amine, compound 2b (Rl=Me, Ra=Me), (140 mg, 0.56 mmol) in DMF (3
mL),
BSTFA (0.59 mL, 2.24 mmol) and triethylamine (0.18 mL, 1.26 mmol) were added
at 0 °C and
then stirred at room temperature for 3 hours. Acid lOb (R9 = butyl) (203 mg,
1.13 mmol) and
89



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
HATU (319 mg, 0.84 mmol) were added and the reaction mixture was stirred for 4
more hours
at room temperature. The DMF was removed and the residue was extracted with
ethyl acetate
(100 mL) and washed with saturated bicarbonate (40 mL). The product obtained
on removal of
solvent was taken up in methanol and treated with Dowex H+ resin for 1 hour.
After filtering the
resin, methanol was removed to obtain the crude product. The product was then
purified on
silica gel column using ethyl acetate as eluent to provide for compound l lb
(Rl=Me, R2=Me
R3=H, R9 = butyl) (200 mg, 86 %).
[0322] 1H NMR (CDC13) 8 8.40 (d, J= 4.2, 1), 8.01 (s, 1), 7.29 (m, 1), 5.40
(d, J = 5.4; 1);
4.02-4.36 (m, 3), 4.80 (s, 1), 3.48-3.60 (m, 1), 3.72 (t, J= 6.0, 2), 2.49 (m,
1}, 2.20 9s, 3), 1.67
(m, 4), 1.40 (m, 3), 0.98-1.18 (m, 9). Mass 413 (M+1).
[0323] To compound 1 lb (Rl=Me, R2=Me R3=H, R9 = butyl) (200 mg, 0. 49 mmol}
in
methanol (5 mL), water (10 mL) and acetic acid (5 mL), platinum dioxide (100
mg, 0.44 mmol)
was added and hydrogenated at 60 psi for 16 h. After filtering the catalyst,
the solvent was
stripped off to obtain the crude product, which was then purified on silica
gel column
chromatography using 20% methanol in dichloromethane as eluent. The lower Rf
fractions
provided the title compound (60 mg, 29%).
[0324] 1H NMR (CDC13): b 5.20 (d, J= 3.6, 1), 4.20 (dd, J= 3.0, 4.8, 1), 4.04
(m, 2), 3.80
(d, J =3.0, 1), 3.61-3.66 (m, l), 3.52 (dd, J= 3.3, 10.2), 2.88 (m, 1), 2.17
(m, 1), 2.14 (s, 3), 1.87
(m, 2), 1.62 (m, 2), 1.32 (m, 6), 0.89 (m, 9); MS (ES+): 419 (M+1).
Example 30
Preparation of 1-(4-cyclohexylpiperid-6-yl)-N ~ 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
iSMe
H



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0325] 4-phenylpyridine-2-carboxylic acid, compound lOb (R9 =phenyl), was made
by
employing general Method P. To 4-phenylpyridine-N oxide (1 g, 5.84 mmol) in
dichloromethane (10 mL) trimethylsilyl cyanide (1.5 mL, 11.6 mmol) and
dimethylcarbamoyl
chloride (1 mL, 11.6 mmol) were added and the reaction mixture was stirred at
room
temperature for 24 hours. An aqueous potassium carbonate solution (10%, 10 mL)
was added
and extracted with dichloromethane (100 mL). The crude product obtained on
removal of
solvent was taken up in hydrochloric acid (6N, 30 mL) and refluxed for 24
hours. Removal of
acid followed by crystallization of the crude product from acetonitrile
resulted in acid lOb (R9' _
phenyl) (1 g, 86%).
[0326] MS (ES-): 198 (M-1); 1H NMR (300 MHz, CD30D) 8 7.64-7.67 (m, 3), 8.02-
8.06
(m, 2), 8.53-8.56 (m, 1), 8.82 (s, 1), 8.82-8.90 (m, 1).
[0327] To the amine 2b (Rl=Me, R~=Me) (102 mg, 0.42 mmol) in DMF (5 mL), BSTFA
(0.549 mL, 2.1 mmol) and triethylamine (0.183 mL, 1.26 mmol) was added at 0
°C and then
stirred at room temperature for 3 hours. Acid lOb (R9~henyl) (158 mg, 0.80
mmol) and HATU
(302 mg, 0.80 mmol) were added and the reaction was stirred for an additional
4 hours at room
temperature. The DMF was removed and the residue was extracted with ethyl
acetate (100 mL)
and washed with saturated bicarbonate (40 mL). The product obtained on removal
of solvent
was taken up in methanol and treated with Dowex H+ resin for 1 hour. After
filtering the resin,
methanol was removed to obtain the crude product. The resulting residue was
then purified by
silica gel chromatography using 10% methanol in dichloromethane as eluent to
provide
compound llb(Rl=Me, R2=Me R3=H, R9' = phenyl) (50 mg, 58 %).
[0328] TLC: Rf = 0.70 (10% MeOH/DCM); MS (ES~: 435 (M+1); 1H NMR (300 MHz,
CDC13) 8 1.01 (t, J = 6.6, 6), 2.12 (s, 3), 2.28 (m, 1), 3.56 (dd, J= 3.3,
10.5, 1), 3.90 (d, J= 3.3,
1), 4.12 (dd, J= 5.4, 10.5, 1), 4.27-4.36 (m, 2), 4.52 (m, 2), 5.26 (d, J=
5.7, 1), 7.48-7.55 (m, 3),
7.77-7.80 (m~ 2), 7.83-7.85 (m, 1), 8.37 (s, 1), 8.69 (d, J= 5.4, 1).
[0329] To compound 1 lb (Rl=Me, Ra=Me R3=H, R9~ = phenyl) (40 mg, 0.09 mmol)
in
methanol (5 mL), water (10 mL) and acetic acid (5 mL), platinum dioxide (100
mg, 0.44 mmol)
was added and the reaction mixture shaken at 60 psi hydrogen for 16 h. The
catalyst was
removed by filtration, and the solvent was evaporated to obtain the crude
product, which was
then purified on silica gel column chromatography using 10% methanol in
dichloromethane to
provide the title compound (10 mg, 25%).
[0330] TLC: Rf= 0.22 (20% MeOH/DCM); MS (ES+): 447 (M+1); 1H NMR (300 MHz,
CD30D) 8 0.90 (d, J= 6.8, 6), 0.93-1.05 (m, 5), 1.20 (m, 6), 1.33-1.47 (m,
4),1.75 (m, 6), 2.10
91



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
(s, 3), 2.18-2.22 (m, 1), 2.97 (t, J= 12.3, 1), 3.39-3.52 (m, 2), 3.70-3.78
(m, 2), 4.05-4.21 (m, 3),
5.23 (d, J= 5.7, 1).
Example 31
Preparation of 1-( 4-ethyl-Nhydroxyethyl-piperid-6-yl)-N { 1-(3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide
6Me
[0331] To the product of Example 1 (28 mg, 0.07 mmol) in methanol (2 mL),
ethylene oxide
(0.5 mL) was added and stirred at 4 °C overnight. The solvent was
removed and the resulting
product was purified by column chromatography using 20% MeOH in DCM as eluent
to obtain
the title compound (16 mg, 51%) as a white powder.
[0332] 1H NMR (300 MHz, CD3OD) b 5.24 (d, J= 6, 1), 4.27 (m, 1), 4.10 (m, 2),
3.95 (m,
1), 3.79-3.50 (m, 4), 3.85 (m, 1) 3.74 (m, 1), 3.26 (m, 1), 2.91 (m, 2), 2.33
(m, 1), 2.13 (m, 4),
1.92 (m, 1), 1.71 (m, 1), 1.17 (m, 7), 0.94 (m, 9); MS (ES+): 435 (M+1).
Example 32
Preparation of 1-( 4-n-pentyl-N hydroxyethyl-piperid-6-yl)-N ~ 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
6Me
92



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0333] The title compound was prepared using the procedures of Example 32 with
the
product from Example 25 as the starting material.
[0334] IH NMR (300 MHz, CD30D) 8 5.24 (d, J--6.0, 1), 4.19 (m, 3), 3.79 (d, J
3.3, 1),
3.74 (m, 1), 3.65 (m, 1), 3.54 (dd, J--3.0, 10.2, 1), 3.25 (m, 2), 2.82 (m,
2), 2.14 (m, 4), 1.89 (m,
1), 1.72 (m, 1), 1.28 (m, 12), 0.94 (m, 9); MS (ES~: 477 (M+1).
Example 33
Preparation of 1-( 4-n-propyl-Nhydroxyethyl-piperid-6-yl)-N { 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
iSMe
H
[0335] The title compound was made using the procedures of Example 32 with the
product
of Example 17 as the starting material.
[0336] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J--6.0, 1), 4.19 (dd, J--9.6; 3.3,
1), 4.11 (m,
2), 3.79 (d, J--3.3, 1), 3.75 (m, 1), 3.65 (m, 1), 3.54 (m, 1), 3.28 (m, 1),
2.82 (m, 2), 2.27 (m, 5)
1.90 (m, 1), 1.71 (m, 1), 1.36 (m, 8), 0.94 (m, 9); MS (ES+): 449 (M+1).
Example 34
Preparation of 1-[4-n-propyl-N (F-moc)-piperid-6-yl]-N { 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-Z-yl]-2-methylprop-1-yl]acetamide
93



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
~ ~Me
io
[0337] To the product of Example 17 (50 mg, 0.123 mmol) in water (3 mL) and
dioxane (3
mL), Fmoc-Cl (38mg, 0.197 mmol) and sodium carbonate (25 mg, 4.246 mmol) were
added and
the reaction mixture was stirred over night at room temperature. The solvents
were removed and
the crude material was loaded into a silica gel column and eluted with ethyl
acetate to obtain the
title compound as a white solid (30 mg, 51%).
[0338] TLC: Rf = 0.5 (EtOAc} MS (ES+) = 627 (M+1), 649 (M+Na); 1H NMR (CD3OD,
200 MHz): 7.79 (d, J= 4.6 Hz, 2), 7.59-7.62 (m, 2), 7.28-7.41 (m, 4), 5.19 (d,
J= 3.8 Hz, 1),
4.45 (m, 2), 4.24 (t, J= 4.2, 1), 3.99-4.15 (m, 4), 3.93 (m, 1), 3.47-3.50 (m,
2), 2.05 (s, 3), 1.87
(m, 1), 1.67 (s, 2), 1.50 (m, 1), 1.30 (m, 4), 0.86-0.91 (m, 9).
Example 35
Preparation of 1-[4-n-propyl-N (carboxylic acid ethyl ester)-piperid-6-yl]-N f
1-[3,4,5
trihydroxy-6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
6Me
)H
[0339] To the product of Example 17 (50 mg, 0.123 mmol) in water (3 mL) and
dioxane (3
mL), ethyl chloroformate (20 mg, 0.147 mmol) and sodium carbonate (25 mg,
0.246 mmol)
were added and stirred over night at room temperature. The solvents were
removed and the
crude material was loaded into a silica gel column and eluted with ethyl
acetate to obtain the title
compound as a white solid (40 mg, 52%).
94



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0340] TLC: Rf = 0.28 (EtOAc). MS (ES~ = 477 (M+1), 499 (M+Na); 1H NMR (CD30D,
200 MHz): 5.22 (d, J= 3.6 Hz, 1), 4.27 (m, 1), 4.03-4.14 (m, 5), 3.96 (bs, 1),
3.62 (m, 1), 3.54
(d, J= 2.2 Hz, 1), 3.52 (d, J= 2.2 Hz, 1), 2.08 (s, 3), 1.93-2.03 (m, 2), 1.75-
1.85 (m, 3), 1.61 (m,
2), 1.33 (m, 4), 1.22-1.28 (m, 3), 0.90-0.94 (m, 9).
Example 36
Preparation of 1-[4-n-propyl-N (carboxylic acid phenyl ester)-piperid-6-yl]-N
~1-[3,4,5
trihydroxy-6-(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl)acetamide
SMe
[0341] To the product of Example 17 (50 mg, 0.123 mmol) in water (3 mL) and
dioxane (3
mL), phenyl chloroformate (40 mg, 0.246 mmol) and sodium carbonate (25 mg,
0.246 mmol)
were added and the reaction mixture was stirred over night at room
temperature. The solvents
were removed and the crude material was loaded into a silica gel column and
eluted with ethyl
acetate to obtain the title compound as a white solid (30 mg, 47%). .
[0342] TLC: Rf = 0.4 (EtOAc). MS (ES+) = 526 (M+1), 548 (M+Na); 1H NMR (CD3OD,
200 MHz): 7.36 (t, J= 3.8 Hz, 2), 7.17-7.23 (m, 10), 7.10 (d, J= 3.6 Hz, 2),
5.20 (d, J= 3.6 Hz,
1), 4.09 (m, 3), 3.93 (d, J= 2.2 Hz, 1), 3.82 (m, 2), 3.46 (m, 2), 2.01 (s,
3), 2.00 (m, 1), 1.71 (m,
1), 1.46-1.36 (m, 4), 0.96-0.90 (m, 9).
Example 37
Preparation of 1-[4- (4,4-difluoropent-1-yl) pyrrolidin-2-yl]-N {1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~acetamide



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
~~?~Ii.
F F NH HNi ~
H ~~~SMe
HO ~H
[0343] To a solution of aldehyde 8a, prepared by first step in general Method
L, (510 mg,
1.47 mmol, 1 equiv) in benzene (8 mL) was added 1-triphenylphosphoranylidene-2-
propanone
(Aldrich) (702 mg, 2.2 mmol, 1.5 equiv). The reaction mixture was refluxed
overnight and the
solvent was removed under vacuum. The residue was purified by chromatography
to give 4-(4-
oxo-pent-2-enyl)-pyrrolidine-1,2-dicarboxylic acid 1-tent-butyl ester 2-methyl
ester as an oil
(237 mg, 42 %).
(0344] MS (ESPOS): 410.2 [M + Na]+, 288.3 [M - Boc + H]+; MS (ESNEG): 386.2 [M
-
H]'.
[0345] To a solution of 4-(4-oxo-pent-2-enyl)-pyrrolidine-1,2-dicarboxylic
acid 1-test-butyl
ester 2-methyl ester (193 mg, 0.5 mmol, l equiv) in benzene (0.9 mL) was added
a solution of
triphenylphosphine-copper (I) hydride hexamer in benzene (3.6 mL). The mixture
was stirred at
rt overnight and hexane (13 mL) was added. The mixture was filtered and the
filtrate was
evaporated. The residue was purified by chromatography to give 4-(4-oxo-
pentyl)-pyrrolidine-
1,2-dicarboxylic acid 1-tent-butyl ester 2-methyl ester (127 mg, 65 %).
[0346] 1H NMR (300 MHz, CDCl3) 8 7.35-7.30 (m, 5), 5.25-5.04 (m, 2), 4.42-4.25
(m, 1),
3.77-3.62 (m, 1), 3.00-2.85 (m, 1), 2.39 (t, J= 7, 2), 2.34-1.47 (m, 7), 2.10
(s, 3), 1.43 (s, 3H),
1.31 (s, 6H); MS (ESPOS): 412.3 [M + Na]+, 290.3 [M - Boc + H]+; MS (ESNEG):
388.4 [M -
H]'.
[0347] To a solution of 4-(4-oxo-pentyl)-pyrrolidine-1,2-dicarboxylic acid 1-
tent-butyl ester
2-methyl ester (155 mg, 0.40 mmol, 1 equiv) in dichloromethane (1.5 mL) at-78
°C was added
DAST (0.21 mL, 1.60 mmol, 4 equiv). The reaction mixture was warmed to rt and
stirred at rt
for 3 h, followed by additional DAST (0.32 mL, 2.4 mmol, 6 equiv) at -78
°C. The mixture was
warmed to rt and stirred overnight. Then the mixture was diluted with
dichloromethane, washed
with sat. aqueous NaHCO3 (lx), dried, and evaporated. The residue was purified
by
chromatography to give 4-(4,4-difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic
acid 1-tent-butyl
ester 2-methyl ester as a yellow oil (88 mg, 54 %).
96



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0348] MS (ESPOS): 434.2 [M + Na]+, 312.3 [M - Boc + H]+.
[0349] To a solution of 4-(4,4-difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic
acid 1-tent-butyl
ester 2-methyl ester (88 mg, 0.21 mmol, 1 equiv) in THF (1.2 mL) and water
(0.4 mL) was
added lithium hydroxide monohydrate (45 mg, 1.07 mmol, 5 equiv). The reaction
mixture was
stirred at rt overnight. The THF was removed under vacuum. The residue was
diluted with
water and washed with ether. The aqueous layer was taken up in ethyl acetate,
partitioned with
10% citric acid. The organic layer was washed with water (1 x), brine (1 x),
dried and
concentrated to give 4-(4,4-difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic acid
1-tent-butyl ester
(66 mg, 96 %).
[0350] 1H NMR (300 MHz, CDCl3) 8 4.39-4.34 (m, 1), 3.57-3.48 (m, 1), 2.92-2.83
(m, 1),
2.57-2.50 (m, 1), 2.30-2.18 (m,l), 1.91-1.73 (m, 3), 1.64-1.36 (m, 7), 1.48
(s, 9); MS (ESPOS):
344.3 [M + Na]+, 222.3 [M - Boc + H]+; MS (ESNEG): 320.2 [M - H]-.
[0351] To a solution of compound 2b (Rl=Me, R2=Me) (50 mg, 0.20 mmol, 1 equiv)
in dry
DMF (0.5 mL) at 0 °C was added triethylamine (88.3 up, 0.64 mmol, 3.2
equiv), followed by the
addition of BSTFA (79.2 ~,L, 0.30 mmol, 1.5 equiv). The reaction mixture was
stirred at 0 °C
for 10 minutes, and then was stirred at rt for 50 minutes. The reaction
mixture was added to the
acid (66 mg, 0.21 mmol, 1 equiv) in a 25 mL round bottom flask, followed by
the addition of
HATU (96.8 mg, 0.25 mmol, 1.25 equiv). The reaction mixture was stirred at rt
for 3 h. The
reaction mixture was evaporated to dryness, taken up in ethyl acetate, washed
with 10% citric
acid, water, sat. NaHC03 and brine. The organic layer was dried over Na2S04
and evaporated
and used in the next step without additional purification.
[0352] To a solution of the above Boc protected lincosamide in DCM (9 mL) with
methyl
sulfide (0.20 mL) were added trifluoroacetic acid (3 mL) and water (0.20 mL).
The reaction
mixture was stirred at rt for 1 h. The solvent was removed under vacuum and co-
evaporated
with toluene twice. The residue was purified by chromatography to provide the
title compound
(68 mg, 75 %) as a white solid.
[0353] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J= 5.4, 1), 4.16 (dd, J= 3.3, 9.9,
1), 4.11-
4.00 (m, 3), 3.75 (d, J= 3.3, 1), 3.51 (dd, J= 3.3, 10.2, 1), 3.40-3.32 (m,
1), 2.71 (dd, J= 8.2,
10.6, 1), 2.23-2.05 (m, 3), 2.10 (s, 3), 1.98-1.76 (m, 3), 1.63-1.39 (m, 7),
0.94-0.87 (m, 6). MS
(ESPOS): 455.3 [M + H] +.
97



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 38
Preparation of 1-[4-(3,3-difluorobut-1-yl)pyrrolidin-2-yl]-N {1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
F F
NH HIS
H ~ ~ iSMe
HO ~H
[0354] Ethyl triphenylphosphonium bromide (Aldrich) (2.92 g, 7.86 mmol, 3.9
equiv)
and potassium-t-butoxide (0.61 g, 5.44 mmol, 2.7 equiv) were suspended in
toluene (26 mL~
under nitrogen with vigorous stirring. After 4 h, a solution of aldehyde 8a
prepared by the first
step in general Method L (700 mg, 2.01 mmol, 1 equiv) in toluene (17 mL) was
added dropwise.
The reaction mixture was stirred at rt for 2 h and diluted with ethyl acetate
(150 mL). The
organic layer was washed with water (2x), brine, dried and concentrated. The
residue was
purified by chromatography to give a clear oil 4-but-2-enyl-pyrrolidine-1,2-
dicarboxylic acid 1-
tert-butyl ester 2-methyl ester (360 mg, 50 %).
[0355] MS (ESPOS): 260.3 [M + H - Boc]+.
[0356] To a solution of 4-but-2-enyl-pyrrolidine-1,2-dicarboxylic acid 1-test-
butyl ester 2-
methyl ester (149 mg, 0.42 mmol, 1 equiv) in DMF (1.4 mL) and water (0.2 mL)
were added
palladium(II)chloride (7.4 mg, 0.042 mmol, 0.1 equiv) and copper(I) chloride
(41.1 mg, 0.42
mmol, 1 equiv). The mixture was stirred at 50 °C overnight with oxygen
bubbling into the
mixture. The mixture was filtered and the filtrate was concentrated under high
vacuum. The
residue was diluted with ethyl acetate, washed with water (lx), brine (lx),
dried and
concentrated. The residue was purified by preparative TLC to give 4-(3-oxo-
butyl)-pyrrolidine-
1,2-dicarboxylic acid 1-tent-butyl ester 2-methyl ester (110 mg, 71 %).
[0357] 1H NMR (300 MHz, CDC13) 8 7.35-7.28 (m, 5), 5.24-5.03 (m, 2), 4.43-4.25
(m, 1),
3.75-3.61 (m, 1), 3.01-2.87 (m, 1), 2.44-2.35 (m,2), 2.28-2.15 (m, 1), 2.11
(s, 3), 2.09-1.98 (m,
1), 1.91-1.51 (m, 3), 1.43 (s, 3.4H), 1.31 (s, 5.6H). MS (ESPOS): 398.3
[M+Na]+, 276.3 [M-
Boc + H]+.
98



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0358] To a solution of the 4-(3-oxo-butyl)-pyrrolidine-1,2-dicarboxylic acid
1-tent-butyl
ester 2-methyl ester (110 mg, 0.29 mmol, 1 equiv) in dichloromethane (1.1 mL)
at-78 °C was
added DAST (0.16 mL, 1.17 mmol, 4 equiv). The reaction mixture was warmed to
rt and stirred
at rt for 3 h, followed by additional DAST (0.23 mL, 1.76 mmol, 6 equiv) at -
78 °C. The
mixture was warmed to rt and stirred overnight. Then the mixture was diluted
with
dichloromethane, washed with sat. aqueous NaHC03 (lx), dried, evaporated. The
residue was
purified by chromatography to give 4-(3,3-Difluoro-butyl)-pyrrolidine-1,2-
dicarboxylic acid 1-
tent-butyl ester 2-methyl ester (92.7 mg, 80 %).
[0359] MS (ESPOS): 420.3 [M + Na]+.
[0360] To a mixture of 4-(3,3-difluoro-butyl)-pyrrolidine-1,2-dicarboxylic
acid 1-test-butyl
ester 2-methyl ester (92.7 mg, 0.23 mmol, 1 equiv) in THF (1.2 mL) and water
(0.4 mL) was
added lithium hydroxide monohydrate (49 mg, 1.17 mmol, 5 equiv). The reaction
mixture was
stirred at rt overnight. the THF was removed under vacuum. The residue was
diluted with
water, washed with ether. The aqueous layer was taken up in ethyl acetate,
partitioned with 10%
citric acid. The organic layer was washed with water (1 x), brine (1 x), dried
and concentrated
to give a white solid, 4-(3,3-difluoro-butyl)-pyrrolidine-1,2-dicarboxylic
acid 1-tent-butyl ester
(59.7 mg, 83 %).
[0361] 1H NMR (300 MHz, CDC13) b 4.40-4.36 (m, 1), 3.59-3.52 (m, 1), 2.94-2.86
(m; 1),
2.55-2.48 (m, 1), 2.33-2.15 (m, 1), 1.92-1.73 (m, 3), 1.66-1.40 (m, 5), 1.47
(s, 9); MS (ESPOS):
330.2 [M + Na]+, 208.2 [M - Boc + H]+; MS (ESNEG): 306.1 [M - H]-.
[0362] To a solution of compound 2b (Rl=Me, R2=Me) (50 mg, 0.20 mmol, 1 equiv)
in dry
DMF (0.5 mL) at 0 °C was added triethylamine (88.3 up, 0.64 mmol, 3.2
equiv), followed by the
addition of BSTFA (79.2 ~L, 0.30 mmol, 1.5 equiv). The reaction mixture was
stirred at 0 °C
for 10 minutes, and then was stirred at rt for 50 minutes. The reaction
mixture was added to the
acid 4-(3,3-difluoro-butyl)-pyrrolidine-1,2-dicarboxylic acid 1-tent-butyl
ester (59.7 mg, 0.20
mmol, 1 equiv) in a 25 mL round bottom flask, followed by the addition of HATU
(93.3 mg,
0.25 mmol, 1.25 equiv). The reaction mixture was stirred at rt for 3 h. The
reaction mixture
was evaporated to dryness, taken up in ethyl acetate, washed with 10% citric
acid, water, s lat.
NaHC03 and brine. The organic layer was dried over Na2S04 and evaporated to
give a syrup.
[0363] To a solution of the above syrup in DCM (9 mL) with methyl sulfide
(0.20 mL) were
added trifluoroacetic acid (3 mL) and water (0.20 mL). The reaction mixture
was stirred at rt for
1 h. The solvent was removed under vacuum and co-evaporated with toluene
twice. The
99



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
residue was purified by chromatography to provide the title compound (63 mg,
72 %) as a white
solid.
[0364] 1H NMR (300 MHz, CD30D) S 5.24 (d, J= 5.7, 1), 4.22-4.13 (m, 2), 4.10-
4.04 (m,
2), 3.76 (d, J= 2.4, 1), 3.54-3.42 (m, 2), 2.84-2.76 (m, 1), 2.29-1.83 (m, 5),
2.10 (s, 3), 1.67-1.51
(m, 6), 0.95-0.87' (m, 6). MS (ESPOS): 441.3 [M + H] +
Example 39
Preparation of 1-[4-(3,3-difluoropent-1-yl)pyrrolidin-2-yl]-N {1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yf~ acetamide
F F
,,. O
NH H
H ~~6Me
HOr~~H
[0365] To a solution of compound 7c (R9'=2-penteneyl) prepared using methods
of general
Method K (323.7 mg, 0.87 mmol, 1 equiv) in DMF (2.8 mL) and water (0.4 mL) at
0 °C were
added palladium (II) chloride (15.4 mg, 0.087 mmol, 0.1 equiv) and copper(I)
chloride (85.9 mg,
0.87 mmol, 1 equiv). The mixture was stirred at 50 °C overnight with
oxygen bubbling into the
mixture. The mixture was filtered and the filtrate was concentrated under high
vacuum. The
residue was diluted with ethyl acetate, washed with water (lx), brine (lx),
dried and
concentrated. The residue was purified by preparative TLC to provide 4-(3-oxo-
pentyl)-
pyrrolidine-1,2-dicarboxylic acid 1-test-butyl ester 2-methyl ester (242 mg,
72 %).
[0366] MS (ESPOS): 412.3 [M + Na]+, 290.3 [M - Boc + H]+.
[0367] To 4-(3-oxo-pentyl)-pyrrolidine-1,2-dicarboxylic acid 1-tent-butyl
ester 2-methyl
ester (242 mg, 0.62 mmol, 1 equiv) in dichloromethane (2.3 mL) at -78
°C was added DAST
(0.33 mL, 2.49 mmol, 4 equiv). The reaction mixture was warmed to rt and
stirred at rt for 3 h,
followed by an addition of more DAST (0.49 mL, 3.73 mmol, 6 equiv) at -78
°C. The mixture
was warmed to rt and stirred overnight. Then the mixture was diluted with
dichloromethane,
washed with sat. aqueous NaHC03 (lx), dried, evaporated. The residue was
purified by
100



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
chromatography to 4-(3,3-Difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic acid 1-
tent-butyl ester
2-methyl ester (117 mg, 46%).
[0368] 1H NMR (300 MHz, CDC13 ) 8 7.35-7.26 (m, 5), 5.25-5.04 (m, 2), 4.44-
4.27 (m, 1),
3.79-3.64 (m, 1), 3.02-2.89 (m, 1), 2.32-2.17 (m,l), 2.13-2.02 (m, 1), 1.91-
1.68 (m, 5), 1.57-1.47
(m, 2), 1.44 (s, 3.SH), 1.31 (s, S.SH), 0.97 (t, J= 7.5, 3).
MS (ESPOS): 434.3 [M + Na]+, 312.3 [M - Boc + H]+.
[0369] To a solution of 4-(3,3-difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic
acid 1-tent-butyl
ester 2-methyl ester (106 mg, 0.26 mmol, l equiv) in THF (2.4 mL) and water
(0.8 mL) was
added lithium hydroxide monohydrate (54 mg, 1.29 mmol, 5 equiv). The reaction
mixture was
stirred at rt overnight. THF was removed under vacuum. The residue was diluted
with water (10
mL), washed with ether (20 mL). The aqueous layer was taken up in ethyl
acetate (50 mL),
partitioned with 10% citric acid (25 mL). The organic layer was washed with
water (1 x), brine
(1 x), dried and concentrated to give 4-(3,3-difluoro-pentyl)-pyrrolidine-1,2-
dicarboxylic acid 1-
tert-butyl ester as a clear oil (82.1 mg, 99%).
[0370] 1H NMR (300 MHz, CDC13) 8 4.40-4.36 (m, 1), 3.58-3.51 (m, 1), 2.94-2.86
(m, 1),
2.57-2.51 (m, 1), 2.30-2.15 (m,l), 1.92-1.72 (m, 5), 1.62-1.53 (m, 2)~, 1.48
(s, 9), 0.99 (t, J= 7.5,
3); MS (ESPOS): 344.3 [M + Na]+, 222.3 [M - Boc + H]+.
[0371] To a solution of compound 2b (Rl=Me, Ra=Me) (50 mg, 0.20 mmol, 1 equiv)
in dry
DMF (0.5 mL) at 0 °C was added triethylamine (88.3 uL, 0.64 mmol, 3.2
equiv), followed by
the addition of BSTFA (79.2 uL, 0.30 mmol, 1.5 equiv). The reaction mixture
was stirred at 0
°C for 10 minutes, and then was stirred at rt for 50 minutes. The
reaction mixture was added to
the acid 4-(3,3-difluoro-pentyl)-pyrrolidine-1,2-dicarboxylic acid 1-tent-
butyl ester (76.6 mg,
0.24 mmol, 1.2 equiv) in a 25 mL round bottom flask, followed by the addition
of HATU (111.9
mg, 0.29 mmol, 1.5 equiv). The reaction mixture was stirred at rt for 3 h. The
reaction mixture
was evaporated to dryness, taken up in ethyl acetate (60 mL), washed with 10%
citric acid (30
mL), water (30 mL), sat. NaHCO3 (30 mL) and brine. The organic layer was dried
over Na2S04
and evaporated to give a yellow oil.
[0372] To a solution of the above oil in DCM (9 mL) with methyl sulfide (0.20
mL)
were added trifluoroacetic acid (3 mL) and water (0.20 mL). The reaction
mixture was stirred at
rt for 1 h. The solvent was removed under vacuum and co-evaporated with
toluene twice. The
residue was purified by chromatography to provide the title compound (72 mg,
80%) as a white
solid.
101



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0373] 1H NMR (300 MHz, CD30D) ~ 5.24 (d, J= 5.7, 1), 4.20-4.04 (m, 4), 3.76
(d, J= 2.7,
1), 3.51 (dd, J= 3.4, 10.3, 1), 3.43 (dd, J= 6.9, 10.8, 1), 2.77 (dd, J= 8.4,
10.8, 1), 2.30-2.05 (m,
3), 2.10 (s, 3), 2.03-1.76 (m, 5), 1.64-1.54 (m, 2), 1.03-0.89 (m, 9); MS
(ESPOS): 455.4 [M + H]
Example 40
Preparation of 1-[4-(3,3-difluoropent-1-yl)-N (2-hydroxyeth-1-yl)pyrrolidin-2-
yl]-l~ ]1-
[3,4,5-trihydroxy-6- (methylthio)tetrahydropyran-2-yl]-2-methylprop-1-
yl]acetamide
F
N H1~ ~
H ~ ~ ~SMe
OH C~--
HO ~OH
(0374] To a solution of the product of Example 42 (17.9 mg, 0.039 mmol) in
MeOH (2 mL)
at 0 °C was added ethylene oxide (0.4 mL). The reaction mixture was
stirred at 4 °C overnight.
The reaction mixture was concentrated and purified by chromatography to give
the title
compound as a white solid (8.2 mg, 42%).
(0375] 1H NMR (300 MHz, CD30D) 8 5.23 (d, J= 5.7, 1), 4.13-4.05 (m, 3), 3.75
(d, J= 3.6,
1), 3.72-3.57 (m, 2), 3.53 (dd, J= 3.3, 10.2, 1), 3.41-3.36 (m, 1), 3.22 (dd,
J= 3.3, 10.8, 1), 2.88-
2.78 (m, 1), 2.63-2.54 (m, 1), 2.18-1.99 (m, 4), 2.10 (s, 3), 1.93-1.75 (m,
5), 1.57-1.46 (m, 2),
1.01-0.90 (m, 9); MS (ESPOS): 499.6 [M + H] +; MS (ESNEG): 497.5 [M - H]-.
Example 41
Preparation of 1-(4-(2,2-difluoroeth-1-yl)piperid-6-yl)-N ~ 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide
102



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
6Me
H
[0376]
[0377] Compound 14c (R9 = 2,2-difluoroethyl) is prepared using the methods
described in
general Method R.
[0378] To a dry flask were added compound 14a (1.4 g, 5.32 mrnol, 1 equiv), ,
triphenylphosphine (111.6 mg, 0.43 mmol, 0.08 equiv), copper (I) iodide (81
mg, 0.43 mmol,
0.08 equiv), palladium acetate (47.7 mg, 0.21 mmol, 0.04 equiv) and
triethylamine (20 mL).
The mixture was deaerated with nitrogen, followed by addition of
propiolaldehyde diethyl acetyl
(1.36 g, 10.65 mmol, 2 equiv). The mixture was stirred at rt for 3 hrs. The
solvent was.removed
under vacuum to give a dark residue. The residue was purified by
chromatography to give a
yellow oil, compound 14b (R9'= 3,3-Diethoxy-prop-1-ynyl) (1.4 g, 100%).
[0379] 1H NMR (300 MHz, CDCl3) 8 8.69 (dd, J= 0.8, 5.0, 1), 8.15 (dd, J= 0.8,
1.4, 1),
7.49 (dd, J=1.7, 5.0, 1), 5.48 (s, 1), 3.99 (s, 3), 3.82-3.73 (m, 2), 3.71-
3.62(m, 2), 1.26 (t, J=
7.2, 6). MS (ESPOS): 264.5 [M + H] +.
[0380] To a solution of 14b (R9'= 3,3-Diethoxy-prop-1-ynyl) (1.4 g, 5.32 mmol)
in methanol
(100 mL) was added 10 % palladium on carbon (0.3 g). The mixture was purged
and charged
with hydrogen (1 atm) and shaken at rt overnight. The palladium was removed by
filtration and
the filtrate was concentrated to give 14c (R9=3,3-diethoxy propyl) as an oil
(1.39 g, 98%).
[0381] 1H NMR (300 MHz, CDC13) ~ 8.60 (d, J= 5.1, 1), 7.98 (d, J= 0.9, 1),
7.31-7.28 (m,
1), 4.45 (t, J= 5.4, 1), 3.98 (s, 3), 3.72-3.58 (m, 2), 3.52-3.39(m, 2), 2.79-
2.72 (m, 2), 1.99-1.90
(m, 2), 1.22-1.15 (m, 6).
[0382] To a mixture of 14c (R9=3,3-diethoxy propyl) (0.68 g, 2.55 mmol) in
acetic acid (8
mL) and water (2 mL) was added conc. hydrochloric acid (2 drops). The mixture
was stirred at
rt overnight and the solvent was removed under high vacuum. The residue was
diluted with
ethyl acetate, washed with sat. sodium bicarbonate (lx), brine (lx). The
organic layer was dried
and concentrated to give 4-(3-Oxo-propyl)-pyridine-2-carboxylic acid methyl
ester as a yellow
oil (0.27 g, 55 %).
103



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0383] To a solution of aldehyde 4-(3-oxo-propyl)-pyridine-2-carboxylic acid
methyl ester
(0.27 g, 1.4 mmol, 1 equiv) in DCM (5 mL) at -78 °C was added DAST
(0.91 g, 5.6 mmol, 4
equiv). The mixture was warmed to rt and stirred overnight. The mixture was
diluted with
dichloromethane (60 mL), washed with sat. aqueous NaHC03 (lx), dried, and
evaporated. The
residue was purified by prep.
[0384] TLC (5% MeOH in DCM) to 4-(3,3-difluoro-propyl)-pyridine-2-carboxylic
acid
methyl ester (137 mg, 45%): 1H NMR (300 MHz, CDC13) 8 8.64 (d, J= 5.1, 1),
8.00-7.98 (m,
1), 7.33-7.29 (m, 1), 5.85 (dddd, J= 4.1, 4.1, 56.4, 56.4, 1), 3.99 (s, 3);
2.90-2.83 (m, 2), 2.28-
2.09 (m, 2); MS (ESPOS): 216.4 [M + H]+.
[0385] To a solution of 4-(3,3-difluoro-propyl)-pyridine-2-carboxylic acid
methyl ester (130
mg, 0.6 mmol) (or compound 14c (R9 = 2,2-difluoroethyl) prepared in the
previous steps) in
MeOH (3 mL} and water (3 mL) were added conc. HCl (0.25 mL, 3.0 mmol, 5 equiv)
and
platinum oxide (65 mg}. The mixture was purged and charged with hydrogen (1
atm) and stirred
overnight. The platinum oxide was removed by filtration and the filtrate was
evaporated to give
a clear syrup. To the above residue were added 2N NaOH (1.21 mL) and t-butyl
alcohol (0.7
mL). The mixture was stirred at rt for 2 hrs. Then di-t-butyl dicarbonate
(0.16 g, 0.73 mmol)
was added. The mixture was stirred at rt overnight. The solvent was removed
under vacuum.
The residue was diluted with water (10 mL), was washed with ether (20 mL). The
aqueous layer
was acidified with 2N HCl to pH = 2.0, and extracted with ethyl acetate (2x).
The combined
organic layers were dried and concentrated to give 4-(3,3-Difluoro-propyl)-
piperidine-1,2-
dicarboxylic acid 1-tent-butyl ester as a clear syrup (163 mg, 88 %)
[0386] 1H NMR (300 MHz, CDC13) ~ 5.77 (dddd, J= 4.2, 4.2, 56.6, 56.6, 1), 4.34
(t, J= 6.4,
1), 3.62-3.50 (m, 1), 3.41-3.30 (m, 1), 2.05-1.96 (m, 1), 1:92-1.73 (m, 4),
1.70-1.60 (m, 1), 1.52-
1.32 (m, 3), 1.43 (s, 9); MS (ESPOS): 330.5 [M + Na] +; MS (ESNEG): 306.5 [M -
H]-.
[0387] To a mixture of the HCl salt of compound 2b (Rl=Me, Ra=Me) (140 mg,
0.49 mmol,
1 equiv) in dry DMF (1.2 mL) at 0 °C was added triethylamine (0.34 mL,
2.43 mmol, 5 equiv),
followed by the addition of BSTFA (0.20 mL, 0.74 mmol, 1.5 equiv). The
reaction mixture was
stirred at 0 °C for 10 minutes, and then was stirred at rt for 50
minutes. To the reaction mixture
were added the 4-(3,3-Difluoro-propyl)-piperidine-1,2-dicarboxylic acid 1-tent-
butyl ester (153
mg, 0.50 mmol, 1.0 equiv) and HATU (235 mg, 0.62 mmol, 1.26 equiv). The
reaction mixture
was stirred at rt for 3 h. The reaction mixture was evaporated to dryness,
taken up in ethyl
acetate, washed with 10% citric acid (lx), water (lx), sat. NaHC03 (lx) and
brine. The organic
104



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
layer was dried over Na2S04 and evaporated to give a pink syrup which was used
without
purification.
[0388] To a solution of the above syrup in DCM (15 mL) with methyl sulfide
(0.33 mL)
were added trifluoroacetic acid (5 mL) and water (0.33 mL). The reaction
mixture was stirred at
rt for 1 h. The solvent was removed under vacuum and co-evaporated with
toluene twice. The
residue was purified by chromatography to provide the title compound (lower
isomer, 93 mg,
43%) as a white solid.
[0389] 1H NMR (300 MHz, CD3OD) 8 5.90 (dddd, J= 4.2, 4.2, 56.7, 56.7, 1), 5.24
(d, J=
6, 1), 4.21 (dd, J= 3.5, 9.8, 1), 4.11-4.04 (m, 2), 3.84-3.77 (m, 2), 3.51
(dd, J= 3.2, 10.3, 1),
3.45-3.37 (m, 1), 3.07-2.98 (m, 1), 2.23-2.12 (m, 2), 2.11 (s, 3)., 1.98-1.66
(m, 4), 1.5.2-1.26 (m,
4), 0.94-0.88 (m, 6). MS (ESPOS): 441.7 [M + H] +.
Example 42
Preparation of 1-(4-(3,3-difluoroprop-1-yl)piperid-6-yl)-N { 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl{ acetamide
~ISMe
H
[0390] To a solution of methyl sulfoxide (0.58 mL, 8.16 mmol, 2.4 equiv) in
dichloromethane ( 1.8 mL) at -72 °C was added a 2 M solution of oxalyl
chloride in
dichloromethane (2.04 mL, 4.08 mmol, 1.2 equiv) over a period of 1 minute. The
mixture was
stirred at -72 °C for 25 minutes, followed by the dropwise addition of
a solution of the alcohol
14c (R9=4-hydroxybutyl), prepaxed using the procedures in general Method R,
(0.71 g, 3.4
mmol, 1 equiv) in dichloromethane (4.8 mL) over a period of 2 minutes. The
reaction mixture
was stirred at -72 °C for 25 minutes, then warmed to -50 °C and
stirred for an additional 2 h.
Triethylamine (1.89 mL, 13.6 mmol, 4.0 equiv) was added and stirred at -50
°C for 25 minutes.
The mixture was diluted with ethyl acetate, washed with water (lx), sat.
aqueous NaHC03 (lx),
105



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
brine (lx), dried, evaporated and co-evaporated with anhydrous toluene to give
aldehyde 4-(4-
oxo-butyl)-pyridine-2-carboxylic acid methyl ester as an oil (0.66 mg, 94%).
(0391] IH NMR (300 MHz, CDC13) 8 9.78 (s, 1), 8.62 (d, J= 5.1, 1), 7.97 (s,
1), 7.29 (d, J=
5.1, 1), 3.99 (s, 3), 2.72 (t, J= 7.8, 2), 2.50 (t, J= 7.2, 2), 2.04-1.93 (m,
2); MS (ESPOS): 230.4
[M + Na]+.
[0392] To a solution of 4-(4-oxo-butyl)-pyridine-2-carboxylic acid methyl
ester (0.66 g, 3.19
mmol, 1 equiv) in DCM (12 mL) at -78 °C was added DAST (1.69 mL, 12.75
mmol, 4 equiv).
The mixture was warmed to rt and stirred overnight. The mixture was diluted
with
dichloromethane, washed with sat. aqueous NaHC03 (lx), brine (lx), dried,
evaporated. The
residue was purified by chromatography to provide 4-(4,4-difluorobutyl)-
pyridine-2-carboxylic
acid methyl ester (0.54 g, 74%).
[0393] 1H NMR (300 MHz, CDC13) ~ 8.62 (d, J= 5.1, 1), 7.97-7.95 (m, 1), 7.29-
7.26 (m, 1),
5.81 (dddd, J= 3.9, 3.9, 56.6, 56.6, 1), 3.98 (s, 3), 2.74 (t, J= 7.2, 2),
1.93-1.77 (m, 4). MS
(ESPOS): 230.4 [M + H]+, 252.4 [M + Na]+.
[0394] To a mixture 4-(4,4-difluorobutyl)-pyridine-2-carboxylic acid methyl
ester (0.54 g,
2.36 mmol, 1 equiv) in MeOH (8 mL) and water (8 mL) were added conc. HCl (0.59
mL, 7.07
mmol, 3 equiv) and platinum oxide (0.2 g). The mixture was purged and charged
with hydrogen
(1 atm) and stirred overnight. The platinum oxide was removed by filtration
and the filtrate was
evaporated to give a residue:
[0395] MS (ESPOS): 236.6 [M + H] +.
[0396] To the residue prepared above were added 2N NaOH (4.72 mL) and t-butyl
alcohol
(2.5 mL). The mixture was stirred at rt for 2 hrs. Then di-t-butyl dicarbonate
(0.77 g, 3.54
mmol) was added. The mixture was stirred at rt overnight. The solvent was
removed under
vacuum. The residue was diluted with water (10 mL), was washed with ether (20
mL). The
aqueous layer was acidified with 2N HCl to pH = 2.0, extracted with ethyl
acetate (2x). The
combined organic layers were dried and concentrated to give 4-(4,4-difluoro-
butyl)-piperidine-1,
2-dicarboxylic acid 1-tent-butyl ester (0.67 g, 89 %).
[0397] ' 1H NMR (300 MHz, CDC13) 8 5.77 (dddd, J= 4.3, 4.3, 56.8, 56.8,1),
4.30 (t, J= 6.8,
1), 3.58-3.47 (m, 1), 3.41-3.31 (m, 1), 2.05-1.96 (m, 1), 1.87-1.68 (m, 4),
1.65-1.56 (m, 1), 1.51-
1.30 (m, 5), 1.43 (s, 9); MS (ESPOS): 344.5 [M + Na] +.
(0398] To a mixture of the HCl salt of compound 2b (Ri=Me, R2=Me) (153 mg,
0.53 mmol,
1 equiv) in dry DMF (1.3 mL) at 0° C was added triethylamine (0.37 mL,
2.66 mmol, 5 equiv),
followed by the addition of BSTFA (0.21 mL, 0.80 mmol, 1.5 equiv). The
reaction mixture was
106



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
stirred at 0° C for 10 minutes, and then was stirred at rt for 50
minutes. To the reaction mixture
were added the 4-(4,4-difluoro-butyl)-piperidine-1,2-dicarboxylic acid 1-tent-
butyl ester (196
mg, 0.61 mmol, 1.15 equiv) and HATU (293 mg, 0.77 mmol, 1.45 equiv). The
reaction mixture
was stirred at rt for 3 h. The reaction mixture was evaporated to dryness,
taken up in ethyl
acetate, washed with 10% citric acid (lx), water (lx), sat. NaHC03 (lx) and
brine. The organic
layer was dried over NaaS04 and evaporated to give a syrup. The residue was
dissolved in
methanol (20 mL), then dried and washed Dowex resin (100 mg) was added. The
mixture was
stirred at rt for 30 minutes, and filtered. The filtrate was concentrated to
give a clear syrup,
which was purified by chromatography to give a clear syrup (0.25g, 85 %).
[0399] MS (ESPOS): 555.8 [M + H] +.
[0400] To a solution of the above syrup in DCM (15 mL) with methyl sulfide
(0.33 mL)
were added trifluoroacetic acid (5 mL) and water (0.33 mL). The reaction
mixture was stirred at
rt for 1 h. The solvent was removed under vacuum and co-evaporated with
toluene twice. The
residue was purified by chromatography to provide the title compound (lower
isomer, 70 mg,
34%) as a white solid.
[0401] 1H NMR (300 MHz, CD30D) 8 5.88 (dddd, J= 4.4, 4.4, 57, 57, 1), 5.24 (d,
J= 5.4,
1), 4.20 (dd, J= 3.2, 10.1, 1), 4.12-4.03 (m, 2), 3.90-3.80 (m, 2), 3.52 (dd,
J= 3.5, 10.3, 1), 3.46-
3.39 (m, 1), 3.09-2.98 (m, 1), 2.25-2.12 (m, 2), 2.11 (s, 3), 1.98-1.67 (m,
4), 1.56-1.30 (m, 6),
0.95-0.87 (m, 6); MS (ESPOS): 455.7 [M + H] +.
Example 43
Preparation of 1-(4-(5,5-difluoropent-1-yl)piperid-6-yl)-N ~ 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
SMe
107



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0402] Method R is used to prepare compound 14c (R9~ = 5-hydroxypentyl) (To a
dry flask
was added compound 14a (2 g, 7.60 mmol, 1 equiv), triphenylphosphine (159.4
mg, 0.61 mrriol,
0.08 equiv), copper (I) iodide (115.8 mg, 0.61 mmol, 0.08 equiv), palladium
acetate (68.2 mg,
0.30 mmol, 0.04 equiv) and triethylamine (28 mL). The mixture was deaerated
with nitrogen,
followed by addition of 4-pentyn-1-of (1.28 g, 15.21 mmol, 2 equiv). The
mixture was stirred at
rt overnight. The solvent was removed under vacuum to give a dark residue. The
residue was
purified by chromatography to give 14b (R9' = 5-hydroxypent-1-yn-yl).
[0403] To a solution of 14b (R9~ _ ~-hydroxypent-1-yn-yl) in methanol (60 mL)
was added
10% palladium on carbon (0.62 g). The mixture was purged and charged with
hydrogen (1 atm)
and stirred at rt overnight. The palladium was removed by filtration and the
filtrate was
concentrated to give a yellow oil 14c (R9 = 5-hydroxypentyl) (1.34 g, 79 %).
[0404] 1H NMR (300 MHz, CDC13) 8 8.62 (d, J= 4.8, 1), 7.97 (s, 1), 7.31 (dd,
J= 1.6, 5, 1),
3.99 (s, 3), 3.63 (t, J= 6.5, 2), 2.70 (t, J= 7.7, 2), 1.74-1.53 (m, 4), 1.46-
1.34 (m, 2).
[0405] To a solution of methyl sulfoxide (0.46 mL, 6.42 mmol, 2.6 equiv)-in
dichloromethane (1.4 mL) at -72 °C was added a 2 M solution of oxalyl
chloride in
dichloromethane (1.61 mL, 3.21 mmol, 1.3 equiv) over a pexiod of 1 minute. The
mixture was
stirred at -72 °C for 25 minutes, followed by the dropwise addition of
a solution of pyridine 14c
(R9 = 5-hydroxypentyl) (0.55 g, 2.47 mmol, 1 equiv) in dichloromethane (3.8
mL) over a period
of 2 minutes. The reaction mixture was stirred at -72 °C for 25
minutes, then warmed to -50 °C
and stirred for an additional 2 h. Triethylamine (1.48 mL, 10.7 mmol, 4.33
equiv) was added
and stirred at -50 °C for 25 minutes. The mixture was diluted with
ethyl acetate, washed with
water (2x), sat. aqueous NaHC03 (lx), brine (lx), dried, evaporated and co-
evaporated with
anhydrous toluene to 4-(5-oxo-pentyl)-pyridine-2-carboxylic acid methyl ester
(0.48 mg, 88 %).
[0406] 1H NMR (300 MHz, CDCl3) 8 9.75 (t, J= 1.4, 1), 8.61 (d, J= 5.1, 1),
7.97-7.95 (m,
1), 7.28 (dd, J= 1.7, 5, 1), 3.99 (s, 3), 2.73-2.67 (m, 2), 2.50-2.44 (m, 2),
1.71-1.63 (m, 4).
[0407] To a solution of the to 4-(5-oxo-pentyl)-pyridine-2-carboxylic acid
methyl ester oil
(0.48 g, 2.19 mmol, 1 equiv) in DCM (8 mL) at -78°C was added DAST
(1.41 g, 8.74 mmol,
4 equiv). The mixture was warmed to rt and stirred overnight. The mixture was
diluted with
dichloromethane, washed with sat. aqueous NaHC03 (lx), dried, and evaporated.
The residue
was purified by chromatography to 4-(5,5-Difluoro-pentyl)-pyridine-2-
carboxylic acid methyl
ester (278 mg, 52 %)
108



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0408] 1H NMR (300 MHz, CDC13) 8 8.61 (dd, J= 0.6, 4.8, 1), 7.97-7.95 (m, 1),
7.28 (dd,
J=1.5, 4.8, 1), 5.78 (dddd, J= 4.3, 4.3, 57, 57, 1), 3.99 (s, 3), 2.70 (t, J=
7.7, 2), 1.94-1.66 (m,
4), 1.55-1.43 (m, 2).
[0409] MS (ESPOS): 244.2 [M + H]+
[0410] To a mixture of 4-(5,5-difluoro-pentyl)-pyridine-2-carboxylic acid
methyl ester (278
mg, 1.14 mmol) in MeOH (5 mL) and water (5 mL) were added conc. HCl (0.286 mL,
3.43
mmol, 3 equiv) and platinum oxide (140 mg). The mixture was purged and charged
with
hydrogen (1 atm) and stirred overnight. The platinum oxide was removed by
filtration and the
filtrate was evaporated to 4-(5,5-difluoro-pentyl)-piperidine-2-carboxylic
acid 2-methyl ester.
[0411] MS (ESPOS): 250.2 [M + H]+.
[0412] To the above residue 4-(5,5-difluoro-pentyl)-piperidine-2-carboxylic
acid-2-methyl
ester were added 2N NaOH (2.3 mL) and t-butyl alcohol (1.2 mL). The mixture
was stirred at rt
for 2 hrs. Then di-t-butyl dicarbonate (0.37 g, 1.72 mmol) was added. The
mixture was stirred
at rt overnight. The solvent was removed under vacuum. The residue was diluted
with water,
was washed with ether. The aqueous layer was acidified with 2N HCl to pH =
2.0, extracted
with ethyl acetate (2x). The combined organic layers were dried and
concentrated to 4-(5,5-
Difluoro-pentyl)-piperidine-1,2-dicarboxylic acid 1-test-butyl ester (310 mg,
81 %).
[0413] 1H NMR (300 MHz, CD30D) 8 5.84 (dddd, J= 4.5, 4.5, 57, 57, 1), 4.31 (t,
J= 6.3,
1), 3.65-3.56 (m, 1), 3.35-3.25 (m, 1), 2.03-1.63 (m, 5), 1.48-1.30 (m, 8),
1.43 (s, 9).
[0414] To a mixture of the HCl salt of compound 2b (RI=Me, R2=Me) (223.7 mg,
0.78
mmol, 1 equiv) in dry DMF (1.9 mL) at 0° C was added triethylamine
(0.54 mL, 3.89 mmol, 5
equiv), followed by the addition of BSTFA (0.31 mL, 1.17 mmol, 1.5 equiv). The
reaction
mixture was stirred at 0° C for 10 minutes, and then was stirred at rt
for 50 minutes. To the
reaction mixture were added 4-(5,5-difluoro-pentyl)-piperidine-1,2-
dicarboxylic acid 1-tert-
butyl ester (272 mg, 0.81 mmol, 1.05 equiv) and HATU (391 mg, 1.03 mmol, 1.32
equiv). The
reaction mixture was stirred at rt for 3 h. The reaction mixture was
evaporated to dryness, taken
up in ethyl acetate, washed with 10% citric acid (lx), water (lx), sat. NaHC03
(lx) and brine.
The organic layer was dried over Na2S04 and evaporated to give a residue. The
residue was
dissolved in methanol (30 mL), then dry and washed Dowex resin (150 mg) was
added. The
mixture was stirred at rt for 1 h and filtered. The filtrate was concentrated
to give a clear syrup,
which was purified by chromatography to give a clear syrup (0.26 g, 72 %).
[0415] To a solution of the above syrup in DCM (15 mL) with methyl sulfide
(0.33 mL)
were added trifluoroacetic acid (5 mL) and water (0.33 mL). The reaction
mixture was stirred at
109



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
rt for 1 h. The solvent was removed under vacuum and co-evaporated with
toluene twice. The
residue was purified by chromatography to provide the title compound (lower
isomer, 40 mg, 15
%) as a white solid.
[0416] 1H NMR (300 MHz, CD30D) 8 5.86 (dddd, J= 4.5, 4.5, 57, 57, 1), 5.24 (d,
J= 5.7,
1 ), 4.21 (dd, J = 3.3, 9.9, 1 ), 4.11-4.04 (m, 2), 3 . 86-3 .78 (m, 2), 3.51
(dd, J = 3 . ~, 10.4, 1 ), 3 .47-
3.38 (m, 1), 3.07-2.97 (m, 1), 2.23-2.12 (m, 2), 2.11 (s, 3), 1.98-1.64 (m,
4), 1.50=1.27 (m, 8),
0.94-0.87 (m, 6); MS (ESPOS): 469.4 [M + H] +.
Example 44
Preparation of 1-(4-(5-fluoropent-1-yl)piperid-6-yl)-N f 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
SMe
[0417] To a solution of compound 14c (R9= 5-hydroxypentyl) (0.66 g, 2.96 mmol,
1 equiv),
prepared as described in general Method R and in the synthesis of Example 47,
in DCM (11 mL)
at -78°C was added DAST (1.91 g, 11.85 mmol, 4 equiv). The mixture was
warmed to rt and
stirred overnight. The mixture was diluted with dichloromethane, washed with
sat. aqueous
NaHC03 (lx), dried, and evaporated. The residue was purified by chromatography
to give 4-(5-
fluoro-pentyl)-pyridine-2-carboxylic acid methyl ester (254 mg, 38 %)
[0418] 1H NMR (300 MHz, CDC13) & 8.62 (d, J= 4.8, 1), 7.97 (d, J= 1.2, 1),
7.30 (dd,
J=1.7, 5, 1), 4.50 (t, J= 5.9, 1), 4.34 (t, J= 6, 1), 3.99 (s, 3), 2.70 (t, J=
7.7, 2), 1.80-1.62 (m,
4), 1.50-1.41 (m, 2).
[0419] To a mixture of 4-(5-fluoro-pentyl)-pyridine-2-carboxylic acid methyl
ester (254 mg,
1.13 mmol) in MeOH (5 mL) and water (5 mL) were added conc. HCl (0.28 mL, 3.39
mmol, 3
equiv) and platinum oxide (130 mg). The mixture was purged and charged with
hydrogen (1
110



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
atm) and stirred overnight. The platinum oxide was removed by filtration and
the filtrate was
evaporated to give 4-(5-fluoro-pentyl)-piperidine-2-carboxylic acid 2-methyl
ester.
[0420] MS (ESPOS): 232.4 [M + H]+.
[0421] To 4-(5-fluoro-pentyl)-piperidine-2-carboxylic acid 2-methyl ester was
added 2N
NaOH (2.43 mL) and t-butyl alcohol (1.3 mL). The mixture was stirred at rt for
2 hrs. Then di-
t-butyl dicarbonate (0.40 g, 1.82 mmol) was added. The mixture was stirred at
rt overnight. The
solvent was removed under vacuum. The residue was diluted with water, was
washed with
ether. The aqueous layer was acidified with 2N HCl to pH = 2.0, extracted with
ethyl acetate
(2x). The combined organic layers were dried and concentrated to give 4-(5-
fluoro-pentyl)-
piperidine-1,2-dicarboxylic acid 1-tent-butyl ester as a syrup (254 mg, 71 %).
[0422] 1H NMR (300 MHz, CDC13) 8 4.52-4.06 (m, 3), 3.55-3.30 (m, 2), 2.03-1.94
(m, 1),
1.81-1.54 (m, 4), 1.45-1.20 (m, 8), 1.43 (s, 9). MS (ESPOS): 218.3 [M + Na -
Boc]'+
[0423] To a mixture of the HCl salt of compound 2b (Rl=Me, R2=Me) (213.8 mg,
0.74
mmol, 1 equiv) in dry DMF (1.8 mL) at 0 °C was added triethylamine
(0.52 mL, 3.72 mmol, 5
equiv), followed by the addition of BSTFA (0.30 mL, 1.12 mmol, 1.5 equiv). The
reaction
mixture was stirred at 0 °C for 10 minutes, and then was stirred at rt
for 50 minutes. To the
reaction mixture were added the 4-(5-fluoro-pentyl)-piperidine-1,2-
dicarboxylic acid 1-tent-butyl
ester as a syrup (244 mg, 0.77 mmol, 1.04 equiv) and HATU (370 mg, 0.97 mmol,
1.31 equiv).
The reaction mixture was stirred at rt for 3 h. The reaction mixture was
evaporated to dryness,
taken up in ethyl acetate, washed with 10% citric acid (lx), water (lx), sat.
NaHC03 (lx) and
brine. The organic layer was dried over Na2S04 and evaporated to give a
residue. The residue
was dissolved in methanol (30 mL), then dry and washed Dowex resin (140 mg)
was added.
The mixture was stirred at rt for 1 h and filtered. The filtrate was
concentrated to give a cleax
syrup, which was purified by chromatography to give a clear syrup (212 mg, 52
%).
[0424] To a solution of the above syrup in DCM (15 mL) with methyl sulfide
(0.33 mL)
were added trifluoroacetic acid (5 mL) and water (0.33 mL). The reaction
mixture was stirred at
rt for 1 h. The solvent was removed under vacuum and co-evaporated with
toluene twice. The
residue was purified by chromatography to provide the title compound (lower
isomer, 40 mg, 17
%) as a white solid.
[0425] 1H NMR (300 MHz, CD30D) 8 5.24 (d, J= 5.7, 1), 4.49 (t, J= 5.9, 1),
4.33 (t, J= 6,
1 ), 4.20 (dd, J = 3.5, 10.1, 1 ), 4.11-4.04 (m, 2), 3 .83-3 .77 (m, 2), 3 .51
(dd, J = 3.3, 10.2, 1 ), 3 .44-
3.36 (m, 1), 3.06-2.94 (m, 1), 2.23-2.13 (m, 2), 2.11 (s, 3), 1.98-1.88 (m,
1), 1.77-1.59 (m, 3),
1.45-1.27 (m, 8), 0.94-0.87 (m, 6).
111



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0426] MS (ESPOS): 451.4 [M + H] +
Example 45
Preparation of 1-(4-(4-fluorobut-1 yl)piperid-6-yl)-N ~ 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl} acetamide
6Me
H
[0427] To a solution of compound 14c (R9 = 4-hydroxybutyl) (0.76 g, 3.62 mmol,
1 equiv),
prepared as described in general Method R and in the synthesis of Example 47,
in DCM (14
mL) at -78 °C was added DAST (1.9 mL, 14.47 mmol, 4 equiv). The mixture
was warmed to rt
and stirred overnight. The mixture was diluted with dichloromethane, washed
with sat. aqueous
NaHC03 (lx), brine (lx), dried, evaporated. The residue was purified by
chromatography to
provide 4-(4-Fluoro-butyl)-pyridine-2-carboxylic acid methyl ester as a yellow
oil (0.24 g, 31
%).
[0428] 1H NMR (300 MHz, CDC13) b 8.57 (d, J= 4.8, 1), 7.92 (d, J= 1.2,1), 7.27-
7.23 (m,
1), 4.49 (t, J= 5.6, 1), 4.33 (t, J= 5.6, 1), 3.94 (s, 3), 2.69 (t, J= 7.5,
2), 1.79-1.59 (m, 4).
[0429] To a mixture of 4-(4-fluoro-butyl)-pyridine-2-carboxylic acid methyl
ester (0.24 g) in
THF (3 mL) and water (1 mL) was added lithium hydroxide monohydrate (71.3 mg,
1.7 mmol,
1.5 equiv). The mixture was stirred at rt overnight and diluted with methanol
(20 mL): Then H+
resin was added and the mixture was shaken for 10 minutes. The resin was
washed with
methanol (lx), 1:1 acetonitrile/water (lx), and acetonitrile (lx). The product
was eluted with
5% TEA in methanol (4x) and acetonitrile (lx). The combined organic solvents
were
evaporated and co-evaporated with toluene to provide 4-(4-Fluoro-butyl)-
pyridine-2-carboxylic
acid (0.22 g, 65 %).
[0430] 1H NMR (300 MHz, CD30D) S 8.46 (d, J= 4.8, 1), 7.95 (s, 1), 7.39-7.35
(m, 1), 4.52
(t, J= 5.6, 1), 4.36 (t, J= 5.9, 1), 3.22 (q, J= 7.3, 2.SH, TEA), 2.77 (t, J=
7.5, 2), 1.84-1.62 (m,
4), 1.28 (t, J= 7.2, 3.8H, TEA).
112



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0431]
[0432] To a solution of 4-(4-fluoro-butyl)-pyridine-2-carboxylic acid (0.22 g,
0.73 mmol, 1
equiv) in dry acetonitrile (4 mL) at 0° C was added triethylamine (74
mg, 0.73 mmol, 1 equiv),
followed by the addition of isobutyl chloroformate (100 mg, 0.73 mmol, 1
equiv). The reaction
mixture was stirred at 0° C for 15 minutes, and then was stirred at
4° C for 2 h. To the reaction
mixture was added a solution of the HCl salt of compound 2b (Rl=Me, Ra=Me)
(263 mg, 0.91
mmol, 1.25 equiv) and triethylamine (93 mg, 0.91 mmol, 1.25 equiv) in a 1:1
acetone/water (4
mL). The reaction mixture was stirred at 4 °C overnight. The reaction
mixture was evaporated
to dryness, taken up in DCM, washed with sat. NaHC03 (lx). The organic layer
was dried over
Na~S04 and evaporated. The residue was purified by chromatography to give a
clear solid (110
mg, 3 5 %).
[0433] To a solution of the above solid (110 mg, 0.25 mmol, 1 equiv) in MeOH
(6 mL) and
water (4 mL) were added cons. HCl (20.2 uL, 0.24 mmol, 0.95 equiv) and
platinum oxide (220
mg). The mixture was purged and charged with hydrogen (65 psi) and shaken
overnight. The
platinum oxide was removed by filtration and the filtrate was evaporated to
give a residue,
which was purified by chromatography to provide the title compound (lower
isomer, 33 mg,
30%) as a white solid.
[0434] 1H NMR (300 MHz, CD30D) b 5.25 (d, J= 5.4, 1), 4.51 (t, J= 6, 1), 4.35
(t, J= 5.9,
1), 4.21 (dd, J= 3.3, 10.2, 1), 4.10-4.04 (m, 2), 3.93-3.80 (m, 2), 3.52 (dd,
J= 3.3, 10.2, 1), 3.46-
3.38 (m, 1), 3.11-2.98 (m, 1), 2.26-2.13 (m, 2), 2.11 (s, 3), 2.00-1.92 (m,
1), 1.80-1.60 (m, 3),
1.54-1.27 (m, 6), 0.95-0.87 (m, 6).
[0435] MS (ESPOS): 437.4 [M + H] +
Example 46
Preparation of 1-(4-(3-ethyl-3-hydroxypent-1-yl)piperid-6-yl)-N { 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~acetamide
113



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
6Me
H
[0436] To a dry flask was added compound 13b (Rl=Me, R2=Me, and R3=H) made
using
general Method Q (130 mg, 0.27 mmol, 1 equiv), triphenylphosphine (45.3 mg,
0.17 mmol, 0.64
equiv), copper (I) iodide (32.9 mg, 0.17 mmol, 0.64 equiv), palladium acetate
(19.4 mg, 0.086
mmol, 0.32 equiv) and triethylamine (1.5 mL). The mixture was deaerated with
nitrogen,
followed by addition of 3-ethyl-1-pentyn-3-of (174 uL, 1.35 mmol, 5 equiv).
The mixture was
stirred at 50 °C overnight. The solvent was removed under vacuum to
give a dark residue. The
residue was purified by chromatography to give 13c (Rl=Me, R2=Me, R3=H, R9=3-
Ethyl-3-
hydroxy-pent-1-ynyl).
[0437] MS (ESPOS): 467.7 [M + H] +; MS (ESNEG): 465.5 [M - H]-.
[0438] To a mixture of the above syrup in MeOH (12 mL) and water (8 mL) were
added
platinum oxide (300 mg) and conc. HCl (26 uL). The mixture was purged and
charged with
hydrogen (65 psi) and shaken overnight. The platinum oxide was removed by
filtration and the
filtrate was evaporated. The residue was purified by chromatography to give
the title compound
as a white solid (19 mg, 15 %).
[0439] 1H NMR (300 MHz, CD30D) b 5.24 (d, J= 5.7, 1), 4.17 (dd, J= 3.1, 10.0,
1), 4.10-
4.02 (m, 2), 3.80 (d, J= 3, 1), 3.53-3.48 (m, 1), 3.42-3.35 (m, 1), 3.23-3.15
(m, 1), 2.75-2.64 (m,
1), 2.22-2.11 (m, 1), 2.10 (s, 3), 2.04-1.97 (m, 1), 1.80-1.72 (m, 1), 1.50-
1.40 (m, 6),1.31-1.06
(m, 5), 0.94-0.80 (m, 12); MS (ESPOS): 477.8 [M + H] +; MS (ESNEG): 475.6 [M -
H]-.
Example 47
Preparation of 1-(4-butoxypiperid-6-yl)-N f 1-[3,4,5-trihydroay-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~acetamide
114



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
NH Hf~ ~~
H ~~~SMe
HO ~OH
[0440] To trimethylsilyl cyanide (5.2 g, 52 mmol), 4-benzyloxy-pyridine 1-
oxide (8.8 g, 44
mmol) in DCM (20 mL) was added, followed by dimethylcarbamoyl chloride (5.6 g,
52 mmol)
in DCM (10 mL), dropwise, stirred at room temperature overnight. Sodium
bicarbonate (100
mL, 10%) was added, stirred for 10 minutes and extracted twice with DCM (50
mL). The
combined organic layer was dried over magnesium sulfate, solvent was removed
to obtain the
product, compound l0a (R9 = Benzyloxy) (10.5 g, 100%).
[0441] 1H NMR (300 MHz, CDC13) 8 8.34 (d, J--5.7, 1), 7.24 (m, 5), 7.11 (t, J--
2.4, 1), 6.90
(dd, J--5.7, 2.4, 1), MS (ES+): 211(M+1).
(0442] Compound l0a (R9 = Benzyloxy) (5 g, 23 mmol) was dissolved in HCl (6N,
70 mL)
and refluxed overnight. The crude product 4-hydroxypyridine-2-carboxylic acid,
compound lOb
(R9 = hydroxy) obtained on removal of HCl was crystallized from acetonitrile
(2.6 g, 80%).
[0443] 1H NMR (300 MHz, CD30D) 8 8.55 (d, J--6.6, 1), 7.78 (d, J--3.0, 1),
6.90 (dd, J--2.7,
6.9, 1), MS (ES-): 138 (M-1).
[0444] The synthesis of title compound was completed using the synthetic
sequence found
in general Method S starting from 4-hydroxypyridine-2-carboxylic acid, as
prepared above.
[0445] 1H NMR (300 MHz, CD3OD) 8 5.25 (d, J 5.4, 1), 4.22 (dd, J--10.2; 3.3,
1), 4.08 (m,
2), 3.81 (d, J--3.0, 1), 3.70 (m, 1), 3.54 (m, 4), 3.43 (m, 2), 2.90 (m, 1),
2.41 (m, 1), 2.19 (m, 1),
2.10 (s, 3) 1.45 (m, 6), 0.92 (m, 9); MS (ES+): 435 (M+1).
115



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 48
Preparation of 1-(4-pentoxypiperid-6-yl)-N { 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl)acetamide
SMe
[0446] The title compound was made using the synthetic sequence found in
general Method
S starting from 4-hydroxypyridine-2-carboxylic acid lOb (R9 = hydroxy)
substituting n-pentyl
bromide as the allcylating agent.
[0447] Compound 15a (Rl° = pentyl): 1H NMR (300 MHz, CD30D) 8 8.38 (d,
J--5.1, 1),
7.64 (s, 1), 7.10 (d, J--3.3, 1), 4.18 (t, J--6.6, 2), 1.85 (m, 2), 1.49 (m,
4), 0.96 (t, J 7.2, 3). MS
(ES-): 208(M-1).
[0448] Compound 15b (Rl=Me, R2=Me, Rl°= butyl): 1H NMR (300 MHz, CD3OD)
8 8.41
(d, J--5.7, 1), 7.61 (d, J 2.4, 1), 7.07 (dd, J--2.4, 5.4, 1), 5.27 (d, J--
5.4, 1), 4.05-4.31 (m, 5), 3.85
(d, J 3.0, 1), 3.57 (dd, J--3.3, 7.2, 1), 2.11 (m, 4), 1.81 (m, 2), 1.49 (m,
4), 1.00 (m, 9). MS
(ES+): 443 (M+1).
[0449] Title compound (20 mg, 10%): 1H NMR (300 MHz, CD30D) b 5.24 (d, J 5.7,
1),
4.22 (dd, J--9.9; 3.3, 1), 4.10 (m, 2), 3.76 (m, 3), 3.51 (m, 3), 3.39 (m, 1),
3.02 (m, 2), 2.43 (m,
1), 2.15 (m, 1), 2.10 (s, 3) 1.95 (m, 2), 1.69 (m, 2), 1.53 (m, 2), 1.34 (m,
2), 0.93 (m, 9); MS
(ES+): 449 (M+1).
116



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 49
Preparation of 1-(4-(4-fluorobutoxy)piperid-6-yl)-N { 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl] acetamide
~SMe
H
[0450] The title compound was made using the synthetic sequence found in
general Method
S starting from 4-hydroxypyridine-2-carboxylic acid lOb (R9 = hydroxy)
substituting 4-
fluorobutyl bromide as the alkylating agent.
[0451] 1H NMR (300 MHz, CD30D) 8 5.25 (d, J--5.7, 1), 4.53 (t, J--5.7, 1),
4.37 (t, J--5.7,
1), 4.21 (dd, J--3.3, 6.6, 1), 4.07 (m, 2), 3.80 (d, J--3.3, 2), 3.60 (m, 5),
2.88 (m, 1), 2.38 (m, 1),
2.18 (m, 1), 2.10 (s, 3) 1.33-1.83 (m, 8), 0.92 (m, 6); MS (ES+): 453 (M+1).
Example 50
Preparation of 1-[4-n-butylprop-1-yl)pyrrolidin-2-yl]-N f 1-[3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-methyl-allyl]acetamide
0
NH HNi ~~
~~~SMe
~H
[0452] To a solution of Boc 7-Methylene MTL (P=Boc, R1=Me, R2=CH2) prepared
from
compound 2a (P=Boc, Rl=Me) by general Method D (391mg, 1.1 mmol) in a solution
of
dichloroethane (10 mL) and dimethylsulfide (0.4 mL, 2.5 mmol) was added, TFA
(5 mL)
117



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
containing water (0.4 mL) and the reaction mixture stirred at rt for 45min.
The solvent was
removed and the residue evaporated twice from DCE to obtain the crude product.
The product
was obtained as an HCl salt by precipitation from ethyl acetate (4 mL) at 0
°C by addition of 2M
HCl in ether, and dried under vacuum (351mg g, 86%). The white solid product
was used in the
next reaction without additional purification.
[0453] MS (ESPOS): 350 (M+H).
[0454] Triethylamine (0.68 mL, 4.9 mmol, 4.0 equiv), followed by BSTFA (0.58
mL, 2.20
mmol, 1.8 equiv), were added to a stirred suspension of compound prepared
above (351 mg,
1.22 mmol, l equiv) in anhydrous DMF (5 mL) at 0 °C and under nitrogen.
The resulting
mixture was stirred at 0 °C for 10 min, and then at rt for 50 min. The
resulting solution was
cooled to 0 °C and a solution of compound 7d (R9=h-butyl) prepared by
Method K (400 mg,
1.47 mmol, 1.2 equiv) in anhydrous DMF (5 mL) was added, followed by solid
HATU (741 mg,
1.95 mmol, 1.6 equiv). The reaction mixture was allowed to warm to rt and
after 2h the reaction
solution was evaporated to dryness under vacuum. The residual oil obtained was
diluted with
EtOAc (200 mL), washed sequentially with 10% citric acid , l :l saturated
aqueous NaHC03,
water, and brine dried over Na2SO4, and evaporated to dryness.
[0455] To a solution of SOmg crude coupling product in 1,2-dichloroethane (6
mL), was
added dimethylsulfide (200 wL), followed by TFA (11.5 mL), and water (768
~,L). The resulting
mixture was stirred at rt for lh, evaporated to a minimal volume, diluted with
DCE (3 x 10 mL),
and evaporated to dryness. The residue was purified by column chromatography
8% to 12%
0.25M methanolic ammonia in dichloromethane to provide the title compound
(10.0 mg, 25 %).
[0456] iH NMR (300 MHz, CD30D) 8 5.22 (d, J= 5.8, 1), 5.00 (s, 1), 4.95 (s,
1), 4.58 (d,
J= 8.8, 1) 4.19 (d, J= 8.8, 1), 4.09 (dd, J=- 5.8, 10.1, 1) 3.85-3.77 (m, 2),
3.57-3.52 (m, 1),
3.26-3.29 (m, 1), 2.59-2.53 (m, 1), 2.10-1.98 (m, 4), 1.80 (s, 3), 1.36-1.51-
1.11 (m, 7), 0.91(t, ,
J--6.9, 3); MS (ESPOS): 403.6 [M + H] +.
118



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 51
Preparation of 1-( 4-ethyl-N ethyl-piperid-6-yl)-N ~ 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
0
,.
O
HO ~ ~ ~SMe
HO ~OH
[0457] To the product of Example 1 (30 mg, 0.07 mmol) in DMF (1 mL), DIEA (43
mg,
0.35 mmol) was added at room temperature and stirred overnight. Then removed
the solvent
and the resulting product was purified by column chromatography using 20% MeOH
in DCM to
obtain the title c~mpound (20 mg, 66%) as a white powder.
[0458] 1H NMR (300 MHz, CD3OD) 8 5.24 (d, J= 6.0, 1), 4.26 (dd, J--3.6, 9.6,
1), 3.79 (d,
J= 3.0, 1), 3.55 (dd, J--3.3, 10.2, 1) 2.85 (m, 2), 2.13 (m, 4), 1.37 (m, 12),
0.94 (m, 9); MS
(ES+): 420 (M+1).
Example 52
Preparation of 1-(4-(3-fluoropropoxy)piperid-6-yl)-N ~ 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
F
O
O
NH HN~
O
HO ~ ~ ~SMe
HO OOH
119



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0459] The title compound was made using the synthetic sequence found in
general
Method S starting from 4-hydroxypyridine-2-carboxylic acid substituting 3-
fluoropropyl
bromide as the alkylating agent.
[0460] Compound 15a (Rl° = 3-fluoropropyl):1H NMR (300 MHz, CD30D) 8
8.41 (d,
J--5.1, 1), 7.65 (d, J--2.1, 1), 7.14 (dd, J--2.1, 5.7, 1), 4.59 (m, 2), 4.24
(t, J--6.0, 2), 1.91 (m, 2).
MS (ES-): 212(M-1).
[0461] Compound 15b (Rl=Me, Ra=Me, Rl° = 3-fluoropropyl): 1H NMR (300
MHz,
CD30D) 8 8.44 (d, J--5.7, 1), 7.65 (d, J--2.4, 1), 7.12 (dd, J--2.4, 5.7, 1};
5.48 (d, J--5.7,1), 4.87
(m, 2), 4.30 (m, 2), 4.12 (dd, J--3.0, 10.2, 1), 3.85 (d, J--3.3, 1}, 3.56
(dd, J--9.9, 3.3, 1), 2.26 (m,
1), 2.11 (s, 3), 1.37 (m, 4), 1.00 (t, J--5.1, 6). MS (ES+): 443 (M+1).
(0462] Title compound (60 mg, 31%): 1H NMR (300 MHz, CD3OD) 8 5.25 (d, J--5.7,
1),
4.50 (m, 2), 4.21 (dd, J 3.3, 9.9, 1), 4.06 (m, 2), 3.80 (d, J--2.7, 1), 3.66
(m, 3), 3.59 (m, 1), 3.33
(m, 1), 2.87 (m, 1), 2.41 (m, 1), 2.18 (m, 1), 2.10 (s, 3) 1.91 (m, 4), 1.51
(m, 2), 0.92 (m, 6); MS
(ES+): 439 (M+1).
Example 53
Preparation of 1-(4-(3,3,3-trifluoropropoxy)piperid-6-yl)-N { 1-[3,4,5-
trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl}acetamide
F
~~~SMe
)H
(0463] The title compound was made using the synthetic sequence found in
general
Method S starting from 4-hydroxypyridine-2-carboxylic acid substituting 2-
trifluoroethyl
bromide as the alkylating agent.
[0464] Compound 15a (Rl° = 2-trifluoroethyl): 1H NMR (300 MHz, CD30D) 8
8.67 (m, 1),
7.92 (s, 1), 7.43 (m, 1), 4.65 (m, 2), 3.01 (m, 2). MS (ES-): 234(M-1).
120



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0465] Compound 15b (Rl=Me, R2=Me, Rl° = 2-trifluoroethyl): 1H NMR (300
MHz,
CD3OD) 8 8.46 (d, J--6.0, 1), 7.65 (d, J--2.7, 1), 7.13 (dd, J--2.7, 6.0, 1),
5.27 (d, J--5.7, 1), 4.39
(t, J--6.0, 2), 4.30 (m, 2), 4.11 (m, 1), 3.85 (d, J 3.0, 1), 3.57 (dd, J--
3.0, 10.2, 1), 2.88 (m, 2),
2.25 (m, 1), 2.11 (s, 3), 1.00 (t, J--6.9, 6). MS (METHOD ES+): 469 (M+1).
[0466] Title compound (10 mg, 10%): 1H NMR (300 MHz, CD3OD) b 5.24 (d, J--5.7,
1),
4.18 (dd, J--3.0, 9.9, 1), 4.15 (m, 2), 3.80 (d, J--3.6, 1), 3.74 (m, 2), 3.52
(dd, J--3.3, 10.2, 2),
3 .3 8 (m, 2), 3.18 (m, 1 ), 2.66 (m, 1 ), 2.66 (m, 1 ), 2.44 (m, 2), 2.22 (m,
1 ), 2.10 (s, 3) 1.34 (m, 2),
0.91 (d, J--7.2, 6); MS (ES+): 475 (M+1).
Example 54
Preparation of 1-(4-iso-butylpiperid-6-yl)-N f 1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~acetamide
0
~ ~ ~SMe
HO~ ~'OH
[0467] 4-Isobutyl-2-cyanopyridine is prepared as follows. To
isobutyltriphenylphosphonium
iodide (Aldrich) (8 g, 18.5 mmol) in THF (20 mL) at 0 °C, potassium-
tert-butoxide (1.8 g, 16
mmol) was added, stirred at room temperature for 1 hr. pyridine-4-
carboxlaldehyde (1 g, 9.3
mmol) was added, stirred at room temperature for overnight. Reaction mixture
was then poured
to water (100 mL) and extracted with EtOAc (100 mL). The product obtained on
removal of
solvent was purified by column chromatography using 40% EtOAc in hexane as
eluent (1.05 g,
84%). To the resulting product (4.2 g, 31.5 mmol), 10% Pd/C (0.4 g) was added
and
hydrogenated at 1 atm pressure overnight. Removal of solvent and purification
on column
chromatography using 30% EtOAc in hexanes resulted in 4-isobutylpyridine (3.8
g, 90%).
[0468] The intermediate, 4-isobutylpyridine-2-carboxylic acid, compound lOb,
(R9 =isobutyl) was made by employing general Method P. To 4-isobutylpyridine
(2 g, 14.8
121



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
mmol) in acetic acid (20 mL), hydrogen peroxide (3.35 g, 30 %, 30 mmol) was
added and
refluxed overnight. After removing the solvent, the residue was dissolved in
DCM dried over
magnesium sulfate and taken as such for the next step. To the compound in DCM
(10 mL)
trimethylsilyl cyanide (1.73 g, 17.4 mmol) and dimethylcarbonyl chloride (1.89
g, 17.4 mmol)
was added and stirred at room temperature for 24 hours. Aqueous potassium
bicarbonate (100
mL, 10%) was added and extracted twice with DCM (50 mL each). The crude
product obtained
on removal of solvent was taken in HCl (6N, 100 mL) and refluxed for 24 hours.
Removal of
acid and purification of crude product by column chromatography using 30% MeOH
in DCM
resulted in acid lOb (R9 =isobutyl) (1.5 g, 100%).
[0469] iH NMR (300 MHz, CD30D) 8 8.78 (d, J--5.7, 1), 8.44 (s, 1), 8.13 (d, J--
5.7, 1), 2.92
(d, J--7.5, 1), 2.15 (m, 1), 0.98 (d, J--6.6, 6). MS (ES-): 178(M-1).
[0470] To the amine 2b (Rl=Me, RZ=Me) (200 mg, 0.79 mmol) in DMF (2m1), TEA
(161
mg, 1.6 mmol), BSTFA (614 mg, 2.4 mmol) was added at 0 °C and stirred
at room temperature
for 1.5 hr. Acid lOb (R9 = isobutyl) (214 mg, 1.2 mmol) and HATU (368 mg, 1.2
mmol) was
added and let stirred at room temperature for 4 hours. DMF was removed and the
residue was
extracted with EtOAc (50 mL), washed with sodium bicarbonate (10%, 50 mL),
brine (50 mL)
and dried over magnesium sulfate. The product obtained on removal of solvent
was dissolved in
methanol (10 mL) and treated with NR-50 resin (300 mg) for 3 hr. After
filtering the resin,
methanol was removed to obtain the crude product. It was then purified on
silica gel column
chromatography using 3% MeOH in DCM to obtain compound llb (Rl=Me, R2=Me,
R3=H, R9
=isobutyl) (200 mg, 60%).
[0471] 1H NMR (300 MHz, CD3OD) 8 8.41 (d, J--4.8, 1), 8:28 (d, J--9.6, 1),
7.95 (s, 1), 5.35
(d, J 5.4, 1), 4.25 (m, 2), 3.99 (d, J 10.8, 1), 3.78 (d, J--3.6, 1), 3.55
(dd, J--3.6, 10.8, 1), 2.52
(m, 3), 2.15 (s, 3), 1.93 (m, 1), 1.02 (m, 12). MS (ES+): 413 (M+1).
[0472] To compound llb (Rl=Me, RZ=Me, R3=H, R9 =isobutyl) (200 mg, 0.48 mmol)
in
water (10 mL), AcOH (2 mL) and MeOH (2 mL), PtO2 (200 mg), was added,
hydrogenated at
55 psi for 16 hours. After filtering the catalyst, the solvent was removed to
obtain the crude
material which on purification over silica gel column using 20 % MeOH in DCM
as eluent. The
lower Rffraction provided the title compound (70 mg, 34%).
[0473] 1H NMR (300 MHz, CD30D) 8 5.25 (d, J--5.7, 1), 4.20 (dd, J--9.9; 3.3,
1), 4.07 (m,
2), 3.80 (d, J--3.0, 1), 3.60 (m, 2), 3.34 (m, 2), 2.84 (m, 1), 2.17 (m, 1),
2.10 (s, 3) 2.01 (m, 1),
1.77 (m, 3), 1.40 (m, 4), 0.91 (m, 12); MS (ES+): 419 (M+1).
122



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 55
Preparation of 1-(4-n-propylpiperid-6-yl)-N f 1-[3,4,5-trihydroxy-6-
(methylthio)tetrahydropyran-2-yl]-2,2-difluoro-prop-1-yl) acetamide
~SMe
)H
[0474] To acid lOb (R9 = propyl) prepared by Method P (53 mg, 0.32 mmol) in
DMF (3
mL), lincosamide intermediate, compound Sb (Rl = Me), prepared by Method I (63
mg, 0.16
mmol) and HATU (121 mg, 0.32 mmol), triethylamine (70 mg, 0.48 mmol) was added
at 0°C
and stirred at room temperature 16 hours. DMF was removed and the residue was
taken in ethyl
acetate and washed with saturated bicarbonate (30 mL). The product obtained on
removal of
solvent was purified on silica gel column using 30% ethyl acetate in hexanes
(40 mg, 45 %). To
the pure product in methanol (5 mL), water (5 mL), acetic acid (5 mL) and
platinum dioxide (50
mg, mmol) was added and hydrogenated at 60 psi for 16 hours. After filtering
the catalyst, the
solvent was removed to obtain the crude product which was taken in methanol (3
mL).
Potassium carbonate (125 mg, 0.83 mmol) in water (1 mL) was added to it and
stirred 16 hours.
Solvents were then removed and the crude product purified on column
chromatography using 20
methanol in dichloromethane. The lower Rf fraction resulted in the title
compound (10 mg,
33%).
[0475] 1H NMR (300 MHz, CD30D) 8 5.28 (d, J= 5.4, 1), 4.73 (s, 1), 4.57-4.65
(m, 1),
4.33-4.42 (m, 1), 4.05 (m, 1), 3.89 (s, 1), 3.53-3.57 (m, 2), 2.83 (t, J=12.3,
1), 2.09 (s, 3), 1.63-
1.84 (m, 5), 1.16-1.37 (m, 6), 0.93 (m, 3). MS(ES+): 427 (M+1).
123



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Example 56
Preparation of 1-[4-n-propyl-4-fluoro-pyrrolidin-2-yl]-N (1-[3,4,5-trihydroxy-
6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl) acetamide
F
O
NH HI~
O
HO ~ ~ ~SMe
HO 'OH
[0476] To a stirred solution of (2S, 4R)-4-hydroxyproline (Aldrich) (25 g, 108
mmol) in
methanol (50 mL) at 0 °C was added trimethylsilyldiazomethene (24.6 g,
216 mmol). The
mixture was stirred at 0 °C for 1 hour. The residue obtained on removal
of solvent and
purification by column chromatography using 50% ethyl acetate in hexanes (27
g, 100%) was
used in the next step. To oxalyl chloride (15 g, 118 mmol) in DCM (15 mL) at -
78 °C, DMSO
(18.6 mL, 236 mmol) was added slowly over 15 minutes. After the completion of
addition, the
above product (2S, 4R)-N-Boc-4-hydroxyproline methylester (26.5 g, 108 mmol)
in DCM (100
mL) was added at -78 °C for 20 minutes. Triethylamine (54.6 g, 540
mmol) was added followed
by stirring at room temperature for 2 hours. The reaction mixture was then
washed with 10% aq
HCl (200 mL) and the organic layer was separated and dried over sodium
sulfate. The crude
product obtained on removal of solvent was purified on silica gel column
chromatography using
50% EtoAc in hexanes to obtain (2S, 4R)-N-Boc-4-Ketoproline methylester (20 g,
78%).
[0477] 1H NMR (300 MHz, CDCl3) 8 4.80 (m, 1), 3.88 (d, J--8.7, 2), 3.77 (s,
3), 2.98 (m, 1),
2.58 (m, 1), 1.45 (s, 9); MS (ES+): 244 (M+1).
[0478] To a stirred solution of (2S, 4R)-N-Boc-4-Ketoproline methylester (1 g,
4.11 mmol)
in THF (10 mL), tetraallyltin (1.08 mL, 4.52 mmol) in dry THF was added, then
cooled to 0 °C
before borontrifluoride etherate (0.520 mL, 4.11 mmol) was added drop wise.
The mixture was
stirred at 0 °C for lh and then at room temperature for an additional 2
hours. Potassium fluoride
(360 mg in SmL water) and celite (1 g) was added and the reaction mixture was
stirred for an
hour. The reaction mixture was filtered and concentrated to dryness and the
residue was
124



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
dissolved in DCM (200 mL), washed with water (100mL) and brine 100 mL), dried
over MgSO4
and evaporated to dryness. T he residue obtained on removal of solvent was
purified by silica
gel column chromatography using 50% EtOAc in hexanes to obtain 4-Hydroxy-4-
allylproline
methylester (0.94 g, 80%).
[0479] 1H NMR (300 MHz, CDC13) 8 5.87 (m, 1), 5.19 (m, 2), 4.34 (m, 1), 3.75
(d, J 4.8,
3), 3.50 (m, 3), 2.37 (m, 1), 2.21 (m, 1), 1.39 (d, J--12.9, 9); MS (ES+): 308
(M+23).
[0480] To a stirred solution of DAST (1.06 g, 6.58 mmol) in DCM (10 mL) at -78
°C,
4-hydroxy-4-allylproline methylester (940 mg, 3.3 mmol) in dry DCM (10 mL) was
added
slowly. The mixture was then stirred at -78 °C for lh, then at -10
°C for an additional lh. DCM
(50 mL) was added, quenched with NH4C1 (.10%, 150 mL) and the organic layer
was separated,
dried over sodium sulfate and evaporated to..dryness. The residue obtained on
removal of solvent
was purified by silica gel column chromatography using 5% EtOAc in hexanes as
eluent to
obtain 4-fluoro-4-allylproline methylester (330 mg, 34%).
[0481] 1H NMR (300 MHz, CDC113) & 5.82 (m, 1), 5.12 (m, 2), 4.43 (m,1), 3.66
(s, 3), 3.47
(m, 1), 2.37 (m, 1), 2.43 (m, 4), 1.37 (dd, J--4.5, 13.8, 9); MS (ES+): 310
(M+23).
[0482] To a solution of 4-fluoro-4-allylproline methylester (0.33 g, 1.15
mmol) in MeOH
(15 mL) was added 10% Pd/C (40 mg) and hydrogenated at 1 atmosphere. The
catalyst was
filtered through celite and washed with methanol. To the product obtained on
removal of solvent
(330 mg, 1.15 mmol) in THF (12 mL) was added aq lithium hydroxide monohydrate
(60 mg,
1.38 mmol). The reaction mixture was stirred at room temperature overnight.
THF was removed
and the residue was taken up in ethyl acetate (50 mL), washed with 10% citric
acid (100 mL)
and brine (20 mL). Removal of solvent resulted in 4-fluoro-4-propylproline
(310 mg, 100%).
[0483] 1H NMR (300 MHz, CD30D) 8 4.43 (m, 1), 3.71 (m, 6), 2.51 (m,.2), 1.98
(m, 3),
1.45 (m, 9), 0.96 (m, 3); MS (ES-): 274 (M-1).
[0484] To a solution of 4-fluoro-4-propylproline (310 mg, 1.15 mmol) in DMF (3
mL) at
0 °C, 7-Methyl MTL 2b (Rl =Me, R2=Me) (272 mg, 1.15 mmol), HBTU (469
mg,~ 1.3 mmol)
and DIEA (290 mg, 2.3 mmol) was added, left stirred at room temperature for 16
hours. DMF
was removed and the residue obtained was purified by 3% MeOH in DCM(40 mg,
93%). The
product from the column purification was taken in DCE (6 mL), to which
triethylsilane (0.16
mL), TFA (2 mL) and water (0.16 mL) was added and stirred at room temperature
for 1.5 hours.
Removal of solvent followed by purification on silica gel column
chromatography using 10%
MeOH in DCM resulted in the title compound as isomeric mixtures with lower RF
fraction (160
mg, 50%).
125



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0485] 1H NMR (300 MHz, CD30D) 8 5.25 (d, J--5.7, 1), 4.46 (m, 1), 4.24 (dd, J-
-5.7, 10.2,
1), 4.08 (m, 2), 3.81 (d, J 2.4, 1), 3.52 (m, 3), 2.73 (m, 1), 2.10 (m, 4),
1.88 (m, 2), 1.50 (m, 2),
0.99 (t, J--7.5, 3), 0.91 (dd, J--3.0, 6.9, 6); MS (ES+): 409 (M+1);) and
higher Rf fraction (40
mg, 12%). 1H NMR (300 MHz, CD30D) 8 5.3 8 (d, .I--5.4, 1 ), 4.46 (m, 1 ), 4.24
(dd, J 2.7, 7.2,
1), 4.08 (m, 2), 3.81 (d, J--2.4, 1), 3.64 (m, 3), 2.73 (m, 1), 2.11 (m, 4),
1.84 (m, 2), 1.47 (m, 2),
0.98 (t, J--7.5, 3), 0.91 (dd, J--3.0, 6.9, 6); MS (METHOD ES+): 409 (M+1).
Example 57
Preparation of 1-[4-n butyl-4-fluoro-pyrrolidin-2-yl]-N (1-[3,4,5-trihydroxy-6
(methylthio)tetrahydropyran-2-yl]-2-methylprop-1-yl~ acetamide
F
O
NH HMw
O
HO ~ ~ ~SMe
HO ~'OH
[0486] To ethyl acetylene (140 mg, 2.6 mmol) in THF (5 mL) at -78 °C, n-
butyllithium (1.1
mL, 2.6 mmol) was added with stirring at -78 °C for 1 hour. Then n-
(tert-Butoxycarbonyl)-L-
proline-4-ketone (described in the example 56) (570 mg, 2.3 mmol) in THF (5
mL) was added at
-78 °C with stirring for 2 hours and then let it warm to -40 °C
over 1 hour. The reaction mixture
was extracted with EtOAc (20 mL), washed with saturated NH4Cl (5 mL) and dried
over sodium
sulfate. Purification of the crude product was carried out by silica gel
chromatography using
50% EtOAc in hexane to obtain the N-boc-4-butyl-4-hydroxy-prolinemethyl ester
(520 mg,
73%). To the DAST (556 mg, 3.4 mmol) in DCM (5 mL) at -78 °C, was added
a solution of the
above ester (520 mg, 1.7 mmol) in DCM (5 mL) at -78 °C and stirred for
1 hour. The reaction
mixture was extracted with DCM (50 mL) and washed with NaHC03 (30 mL, 10%).
The
product obtained after removal of solvent was purified by silica gel
chromatography using 5%
EtOAc in hexanes to obtain N-(tert-butoxycarbonyl)-L-proline-4-fluoro-4-butane
(276 mg,
52%).
126



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0487] 1H NMR (300 MHz, CD30D) 8 4.41 (m, 1), 3.83 (m, 1), 3.71 (s, 3), 3.45
(m, 1),
2.55-1.54 (m, 8), 1.39 (m, 9), 0.89 (m, 3); MS (ES+): 326 (M+23).
[0488] To a solution of N-(tert-butoxycarbonyl)-L-proline-4-fluoro-4-butane
(270 mg, 0.89
mmol) in THF (12 mL) and water (4 mL), was added lithium hydroxide monohydrate
(45 mg,
1.07 mmol). The reaction mixture was stirred at room temperature for 16 hours.
THF was
removed under vacuum and the residue was taken up in ethyl acetate (150 mL),
washed with
10% citric acid (100 mL} and brine (20mL). Removal of solvent resulted in N-
(tert-butoxy)-L-
proline-4-fluoro-4-butyl-2-carboxylic acid (260 mg, 100%).
[0489] 1H NMR (300 MHz, CD30D) 8 4.32 (m, 1), 3.72 (m, 2), 2.5.8 (m, 2), 2.10-
1.63 (m,
6), 1.42 (m, 9}, 0.93 (t, J--6.6, 3); MS (ES-): 288 (M-1).
[0490] To a solution of N-(tert-butoxy)-L-proline-4-fluoro-4-butyl-2-
carboxylic acid (135
mg, 0.46 mmol) in DMF (3 mL) at 0 °C, 7-Methy MTL 2b (Rl =Me, RZ=Me)
(135 mg, 0.46
mmol), HBTU (194 mg, 0.51 mmol), DIEA (120 mg, 0.93 mmol) was added, left it
at room
temperature for 16 hours. The product obtained after removing DMF and
purification by column
chromatography using 5% MeOH in DCM (189 mg, 77%) was taken in DCE (6 mL).
Triethylsilane (0.16 mL), TFA (2 mL) and water (0.16 mL) was added, stirred at
room
temperature for 1.5 hours. The residue obtained on removal of solvent was
purified by column
chromatography using 10% MeOH in DCM to obtain the title compound (156 mg,
96%).
[0491] 1H NMR (300 MHz, CD30D) 8 5.26 (d, J 5.7, 1), 4.55 (m, 1), 4.27 (dd, J
3.3, 10.2,
1), 4.08 (m, 2), 3.82 (d, J--3.0, 1), 3.58 (m, 3), 2.79 (m, 1), 2.22 (m, 1),
2.10 (s, 3), 1.89 (m, 3),
1.40 (m, 4), 0.91 (m, 9); MS (ES+): 423 (M+1)..
Example 58
4-Fluoro-4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxy-1-(3,4,5-trihydroxy-
6-methylsulfanyl
tetrahydro-pyran-2-yl)-propyl]-amide
F
H H~ ~. OH
H ~~ ~SMe
HO~~OH
127



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0492] N-(tent-butoxycarbonyl)-4-fluoro-4-propyl-L-proline was prepared as
described in
the previous example (except using n-propyl lithium in the place of n-butyl
lithium) (164 mg,
0.57 mmol) was suspended in dry acetonitrile (4 mL). Triethylamine (332 ~.L,
3.02 mmol) was
added and the reaction mixture was cooled to 0°C. Isobutyl
chloroformate (78 ~L, 0.57 mmol)
was added and after 10 min the reaction was allowed to warm to 4°C.
After 1.5 h a solution of
MTL (151 mg, 0.57 mmol) in 1:1 acetone: water (4 mL) was added and the
reaction mixture was
stirred for l Oh at rt. The reaction mixture was evaporated to dryness and
chromatographed on
silica 95:5 dichloromethane/MeOH to 95:8 dichloromethane/MeOH to provide the
product as a
colorless oil (137 mg, 45%): TLC Rf 0.32 (9:1 dichloromethane/MeOH).
[0493] To a solution of the above boc protected lincosamide (125 mg,) in DCM
(2.0 mL)
was added a solution of DCE (10.0 mL), trifluoroacetic acid (5 mL) methyl
sulfide (0.3 mL);
and water (0.3 mL). The reaction mixture was stirred at rt for 40 min then
diluted with DCE
(25.0 mL). The solvent was removed under vacuum and co-evaporated with DCE
twice. The
residue was purified by chromatography on fluorosil 20% MeOH (0.25M NH3) in
DCM to
provide the product as a colorless solid (30.0 mg, 30%).
Example 59
Preparation of 4-propyl-pyrrolidine-2-carboxylic acid [2-hydroxy-1-(3,4,5-
trihydroxy-6
methylsulfanyl-tetrahydro-pyran-2-yl)-propyl]-amide
6Me
[0494] The title compound was made using the synthetic sequence found in
general
Method S starting from 4-hydroxypyridine-2-carboxylic acid, substituting 2-
methoxyethyl
bromide as the alkylating agent.
[0495] Compound 15a (Rl° = 2-methoxyethyl): 1H NMR (300 MHz, CD3OD) S
8.40 (d,
J 6.0, 1), 7.69 (d, J--2.4, 1), 7.20 (dd, J--2.7, 6.3, 1), 4.35 (m, 2), 3.80
(m, 2), 3.40 (s, 3). MS
(ES'): 196(M-1).
128



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
[0496] Compound 15b (Rl=Me, Ra=Me, Rl°= 2-methoxyethyl): 1H NMR (300
MHz,
CD30D) 8 8.43 (d, J--5.7, 1), 7.65 (d, J--2.4, 1), 7.12 (dd, J--2.4, 5.7, 1),
5.27 (d, J--5.4, 1), 4.10-
4.87 (m, 4), 3.85 (d, J--3.3, 1), 3.77 (m, 2), 3.55 (m, 1), 3.41 (s, 3), 2.26
(m, 1), 2.11 (s, 1), 0.998
(m, 6). MS (ES~: 431 (M+1).
[0497] Title compound (10 mg, 10%). 1H NMR (300 MHz, D20) 8 5.18 (d, J 6.0,
1), 4.00
(m, 3), 3.70 (m, 1), 3.56 (m, 1), 3.45 (m, 3), 3.26 (m, 1), 3.16 (m, 3), 3.10
(m, 1), 2.80 (m, 1),
2.48 (m, 1), 2.22 (m, 1), 1.96 (m, 4), 1.17-1.72 (m, 4), 0.70 (m, 6); MS
(ES+): 437 (M+1).
[0498] Examples 60 to 65, as indicated in Table 1 above, were prepared using
methods and
techniques describe herein utilizing commercially available starting materials
where appropriate.
[0499] The following Examples may be used to test compounds of this invention.
Example A
Susceptibility Testing
[0500] Compounds were tested following the microdilution method of NCCLS
(National
Committee for Clinical Laboratory Standards. Methods for dilution
antimicrobial susceptibility
tests for bacteria that grow aerobically; Approved standard - fifth edition.
NCCLS document
M7-A5, NCCLS, Wayne, PA. 2000; National Committee for Clinical Laboratory
Standards.
Methods for antimicrobial susceptibility testing of anaerobic bacteria;
Approved standard - fifth
edition. NCCLS document M11-A4, NCCLS, Wayne, PA. 2001). Assays were performed
in
sterile plastic 96-well microtiter trays with round bottom wells (Greiner).
Compound Preparation
[0501] Stock solutions of test compounds and control antibiotics are prepared
at l Omg/mL
in DMSO. Serial 2-fold dilutions of each drug are performed in a microtiter
plate across each
row using DMSO as solvent at 100- fold the desired final concentration. Wells
in columns #1-
11 contain drug and column #12 was kept as a growth control for the organism
with no drug.
Each well in the mother plate is diluted with sterile deionized water, mixed,
and volumes of 10
~L distributed to each well in the resulting assay plates.
Preparation of Inoculum
(0502] Stock cultures were prepared using the MicrobankTM method (Pro-Lab
Diagnostics)
and stored at -80°C. To propagate aerobic strains, one bead was removed
from the frozen vial
129



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
and aseptically streaked onto Trypticase Soy Agar (Difco), Chocolate Agar
(Remel) or Blood
Agar (Remel) which were incubated at 35°C overnight. Anaerobes were
cultivated in Brucella
Agar (Remel) supplemented with hemin and vitamin K and incubated in
anaerobiosis using an
Anaerobic Jar (Mitsubishi) at 35°C for 24 to 48 h. Standardized inocula
were prepared using the
direct colony suspension method according to NCCLS guidelines (National
Committee for
Clinical Laboratory Standards. Methods for dilution antimicrobial
susceptibility tests for
bacteria that grow aerobically; Approved standard - fifth edition. NCCLS
document M7-A5,
NCCLS, Wayne, PA. 2000; National Committee for Clinical Laboratory Standards.
Methods
for antimicrobial susceptibility testing of anaerobic bacteria; Approved
standard - fifth edition.
NCCLS document M11-A4, NCCLS, Wayne, PA. 2001}. Isolated colonies were
selected from
an 18-24 h agar plate and resuspended in 0.9% sterile saline to match a 0.5
McFarland turbidity
standard. The suspension was used within 15 min. of preparation.
Streptococcus pneumoniae VSPN1001 Streptococcus pneumoniae ATCC 49619
Streptococcus pheumohiae VSPN3026Streptococcus prceumoniae R6x


Streptococcus prteumoniae Streptococcus paeumoniae 488K
VSPN4054


Streptococcus pneumohiae VSPN4021Streptococcus prceumoniae 9


Staphylococcus aureus VSAU1017Staphylococcus aureus Smith


Staphylococcus aureus VSAU1003Staphylococcus aureus ATCC 25923


Staphylococcus aureus VSAU4020Staphylococcus aureus125


Staphylococcus aureus VSAU4048Staphylococcus aureus 85-EPI


Staphylococcus aureus VSAU4065Staphylococcus aureus VSAU4065


Staphylococcus epidermidis Staphylococcus epidermidis ATCC
VSEP1001 12228


Enterococcus faecalis VEFL1003Ercterococcus faecalis ATCC
51299


Enterococcus faecium VEFA1005Er~terococcus faecium BM4147.1


Haemophilus ihfuerczae VH1N1003Haemophilus ihfuenzae ATCC 49766


Haemophilus ircfuerczae VH1N1004Haemophilus ihfuenzae ATCC 31517


Haemophilus ihfuenzae VHIN1005Haemophilus ihfuehzae LS-2
acr


Horaxella catarrhalis VMCA1001Horaxella catarrhalis ATCC 25238


Escherichia coli VECO2096 Escherichia coli MG1655


Escherichia coli VEC02526 Escherichia coli MG1655 tolC
tolC


Bacteroides fragilis VBFR1001Bacteroides fragilis ATCC 25285


Bacteroides thetaiotaomicron Bacteroides thetaiotaomicron
VBTH 1001 ATCC #29741


Clostridium difficile VCDI1001Clostridium docile ATCC 9689


130



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Preparation of Assa~Plates for MICs Preparation of Assay Plates for MICs
[0503] Media were prepared at 1.1 x concentration. Mueller-Hinton Broth MHB
(Difco)
supplemented with Ca++ and Mg++ as recommended by NCCLS, MHB supplemented with
5%
horse lysed blood, HTM Broth (Remel), or Brucella broth (Remel) supplemented
with hemin
and vitamin K. For each organism, the standardized suspension was diluted into
appropriate
growth medium in a sterile reservoir. After mixing, wells in the drug-
containing assay plates
were inoculated with a volume of 90p,1. Thus, for each MIC determination, each
well contains a
final volume of 100 ~L with an inoculum size of approximately 5 * 105 cfu/mL
and no more
than 1% DMSO.
Intemretation of MIC
[0504] The completed microtiter plates were incubated 16-20 h at 35°C
in ambient air for
aerobes, and at 35 °C for 46-48 h or in an anaerobe jar (Mitsubishi)
for anaerobes. Optical
density of each well was determined at 600 nm using a VersaMax Microplate
reader (Molecular
Devices, Sunnyvale, CA). The MIC was defined as the lowest drug concentration
causing
complete suppression of visible bacterial growth.
Example B
Efficacy in Marine S. aureus Septicemia
[0505] Efficacy studies were performed in an S. aureus marine septicemia model
according
to models published elsewhere (Goldstein, B. P., G. Candiani, T. M. Arain, G.
Romano, I.
Ciciliato, M. Berti, M. Abbondi, R. Scotti, M. Mainini, F. Ripamonti, and et
al. 1995.
Antimicrobial activity of MDL 63,246, a new semisynthetic glycopeptide
antibiotic Antimicrob
Agents Chemother. 39:1580-1588.; Misiek, M., T. A. Pursiano, F. Leitner, and
K. E. Price 1973.
Microbiological properties of a new cephalosporin, BL-S 339: 7-
(phenylacetimidoyl-
aminoacetamido)-3-(2-methyl-1,3,4-thiadiazol-5-ylthio methyl)ceph-3-em-4-
carboxylic acid
Antimicrob Agents Chemother. 3:40-48).
Compound Preparation
[0506] Compounds were dissolved in 2% Tween 80 for oral dosing or 0.9% NaCI
solution
for intravenous dosing. Compounds were administered at 1 hour after bacterial
inoculation.
Vancomycin or ampicillin were used as controls.
131



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
Efficacy model
[0507] Male or female ICR mice weighing 222 g from MDS Pharma Services were
used
for the evaluation. Food and water was given ad libitum. Groups of 6 mice
weighing 22 ~ g
were used for the experiment. Mice were inoculated intraperitoneally with
Staphylococcus
aureus Smith at 4 104 CFU in 0.5 mL of Brain Heart Infusion Broth (Difco)
containing 5%
mucin (Sigma). Mortality was recorded once daily for 7 days following
bacterial inoculation.
[0508] While the invention has been described and illustrated herein by
references to various
specific material, procedures and examples, it is understood that the
invention is not restricted to
the particular material combinations of material, and procedures selected for
that purpose.
Numerous variations of such details can be implied as will be appreciated by
those skilled in the
art.
Example C
In vivo animal model
[0509] In vivo activity of various compounds of the subject invention was
evaluated in a
standard Staphylococcus aureus septicemia model (MDS Pharma Services, Bothell,
WA).
[0510] Male ICR-derived mice (ICR is a strain of out-bred mice) provided by
MDS Pharma
Services animal breeding center were inoculated intraperitonealy with LD9o_ioo
of
Staphylococcus aureus (Smith; ATCC19636) in O.SmL BHI broth containing 5%
mucin
(Sigma). Compounds were formulated in 2% Tween 80 (Sigma) and single doses
were
administered orally one hour after bacterial inoculation. Mortality was
monitored daily for
seven days.
[0511] In previous studies, the oral EDso (i. e., concentration that protected
50% of the mice)
was determined to be 19.9 mg/kg for clindamycin, a commercially available
lincosamide
(Sigma). To screen the compounds of this invention that were tested, compounds
were
administered at 10 mg/kg to a group of eight ICR mice and the number of
survivors at that
concentration was compared to clindamycin. Results are presented in the table
below.
Compound TestedNo. of surviving
mice


(Indicated by at 10 mg/kg


Example No.)


Clindamycin 6


17 1


34 1


132



CA 02493799 2005-O1-26
WO 2004/016632 PCT/US2003/025820
35 2


35 0


It is assumed that when compound from example 35 is tested at a higher dose,
the
number of surviving mice would increase.
133

Representative Drawing

Sorry, the representative drawing for patent document number 2493799 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2003-08-15
(87) PCT Publication Date 2004-02-26
(85) National Entry 2005-01-26
Examination Requested 2008-08-06
Dead Application 2011-08-15

Abandonment History

Abandonment Date Reason Reinstatement Date
2010-08-16 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2005-01-26
Registration of a document - section 124 $100.00 2005-04-15
Registration of a document - section 124 $100.00 2005-04-15
Maintenance Fee - Application - New Act 2 2005-08-15 $100.00 2005-07-11
Maintenance Fee - Application - New Act 3 2006-08-15 $100.00 2006-07-20
Maintenance Fee - Application - New Act 4 2007-08-15 $100.00 2007-07-23
Maintenance Fee - Application - New Act 5 2008-08-15 $200.00 2008-06-27
Request for Examination $800.00 2008-08-06
Maintenance Fee - Application - New Act 6 2009-08-17 $200.00 2009-06-25
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
VICURON PHARMACEUTICALS, INC.
Past Owners on Record
ANANDAN, SAMPATH K.
GORDEEV, MIKHAIL F.
LEWIS, JASON
PATEL, DINESH V.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2005-01-26 133 6,892
Claims 2005-01-26 11 442
Abstract 2005-01-26 1 54
Cover Page 2005-04-01 1 29
Prosecution-Amendment 2008-08-06 1 31
PCT 2005-01-26 18 792
Assignment 2005-01-26 3 91
Correspondence 2005-03-30 1 27
Assignment 2005-04-15 7 311
PCT 2005-01-27 3 140